WO2006066078A2 - Anticorps specifiques de fc$g(g)riib et leurs procedes d'utilisation - Google Patents

Anticorps specifiques de fc$g(g)riib et leurs procedes d'utilisation Download PDF

Info

Publication number
WO2006066078A2
WO2006066078A2 PCT/US2005/045586 US2005045586W WO2006066078A2 WO 2006066078 A2 WO2006066078 A2 WO 2006066078A2 US 2005045586 W US2005045586 W US 2005045586W WO 2006066078 A2 WO2006066078 A2 WO 2006066078A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
antibodies
fcγriib
fragment
cells
Prior art date
Application number
PCT/US2005/045586
Other languages
English (en)
Other versions
WO2006066078A3 (fr
Inventor
Scott Koenig
Maria Concetta Veri
Nadine Tuaillon
Original Assignee
Magrogenics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Magrogenics, Inc. filed Critical Magrogenics, Inc.
Priority to CA2590935A priority Critical patent/CA2590935C/fr
Priority to EP05854332A priority patent/EP1833849A4/fr
Publication of WO2006066078A2 publication Critical patent/WO2006066078A2/fr
Publication of WO2006066078A3 publication Critical patent/WO2006066078A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against Fc-receptors, e.g. CD16, CD32, CD64
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to antibodies or fragments thereof that specifically bind the extracellular domain of Fc ⁇ RIIB, particularly human Fc ⁇ RIIB, and block the Fc binding site of human Fc ⁇ RIIB.
  • the invention provides methods of treating cancer and/or regulating immune complex mediated cell activation by administering the antibodies of the invention to enhance an immune response.
  • the invention also provides methods of breaking tolerance to an antigen by administering an antigen-antibody complex and an antibody of the invention.
  • the Fc receptors members of the immunoglobulin gene superfamily of proteins, are surface glycoproteins that can bind the Fc portion of immunoglobulin molecules. Each member of the family recognizes immunoglobulins of one or more isotypes through a recognition domain on the a chain of the Fc receptor. Fc receptors are defined by their specificity for immunoglobulin subtypes. Fc receptors for IgG are referred to as Fc ⁇ R, for IgE as Fc ⁇ R, and for IgA as Fc ⁇ R.
  • Different accessory cells bear Fc receptors for antibodies of different isotype, and the isotype of the antibody determines which accessory cells will be engaged in a given response (reviewed by Ravetch J.V. et al. 1991, Annu. Rev. Immunol. 9: 457-
  • Each member of this family is an integral membrane glycoprotein, possessing extracellular domains related to a C2-set of immunoglobulin-related domains, a single membrane spanning domain and an intracytoplasmic domain of variable length.
  • Fc ⁇ Rs There are three known Fc ⁇ Rs, designated Fc ⁇ RI(CD64), Fc ⁇ RII(CD32), and Fc ⁇ RIII(CD16).
  • the three receptors are encoded by distinct genes; however, the extensive homology between the three family members suggest they arose from a common progenitor perhaps by gene duplication. This invention specifically focuses on Fc ⁇ RII(CD32).
  • Fc ⁇ RII proteins are 40KDa integral membrane glycoproteins which bind only the complexed IgG due to a low affinity for monomeric Ig (10 6 M "1 ). This receptor is the most widely expressed Fc ⁇ R, present on all hematopoietic cells, including monocytes, macrophages, B cells, NK cells, neutrophils, mast cells, and platelets. Fc ⁇ RII has only two immunoglobulin- like regions in its immunoglobulin binding chain and hence a much lower affinity for IgG than Fc ⁇ RI. There are three human Fc ⁇ RII genes (Fc ⁇ RII- A, Fc ⁇ RII-B, Fc ⁇ RII-C), all of which bind IgG in aggregates or immune complexes.
  • ITIMS immunoreceptor tyrosine based inhibitory motifs
  • ITAM phosphorylation serves as a docking site for Syk kinase, activation of which results in activation of downstream substrates (e.g., PI 3 K). Cellular activation leads to release of proinflammatory mediators.
  • downstream substrates e.g., PI 3 K.
  • Cellular activation leads to release of proinflammatory mediators.
  • a neoplasm, or tumor is a neoplastic mass resulting from abnormal uncontrolled cell growth which can be benign or malignant. Benign tumors generally remain localized. Malignant tumors are collectively termed cancers.
  • malignant generally means that the tumor can invade and destroy neighboring body structures and spread to distant sites to cause death (for review, see Robbins and Angell, 1976, Basic Pathology, 2d Ed., W.B. Saunders Co., Philadelphia, pp. 68-122). Cancer can arise in many sites of the body and behave differently depending upon its origin. Cancerous cells destroy the part of the body in which they originate and then spread to other part(s) of the body where they start new growth and cause more destruction.
  • Lung and prostate cancer are the top cancer killers for men in the United States.
  • Lung and breast cancer are the top cancer killers for women in the United States.
  • One in two men in the United States will be diagnosed with cancer at some time during his lifetime.
  • One in three women in the United States will be diagnosed with cancer at some time during her lifetime.
  • cancer therapy may involve surgery, chemotherapy, hormonal therapy and/or radiation treatment to eradicate neoplastic cells in a patient (See, for example, Stockdale, 1998, "Principles of Cancer Patient Management", in Scientific American: Medicine, vol. 3, Rubens tein and Federman, eds., Chapter 12, Section IV).
  • cancer therapy could also involve biological therapy or immunotherapy. All of these approaches pose significant drawbacks for the patient.
  • Surgery for example, may be contraindicated due to the health of the patient or may be unacceptable to the patient. Additionally, surgery may not completely remove the neoplastic tissue.
  • Radiation therapy is only effective when the neoplastic tissue exhibits a higher sensitivity to radiation than normal tissue, and radiation therapy can also often elicit serious side effects. Hormonal therapy is rarely given as a single agent and although can be
  • NYJD: 1603354.2 effective is often used to prevent or delay recurrence of cancer after other treatments have removed the majority of the cancer cells.
  • Biological therapies/immunotherapies are limited in number and may produce side effects such as rashes or swellings, flu-like symptoms, including fever, chills and fatigue, digestive tract problems or allergic reactions.
  • chemotherapy there are a variety of chemotherapeutic agents available for treatment of cancer.
  • a significant majority of cancer chemotherapeutics act by inhibiting DNA synthesis, either directly, or indirectly by inhibiting the biosynthesis of the deoxyribonucleotide triphosphate precursors, to prevent DNA replication and concomitant cell division (See, for example, Gilman et ah, Goodman and Gilman's: The Pharmacological Basis of Therapeutics, Eighth Ed. (Pergamom Press, New York, 1990)).
  • agents which include alkylating agents, such as nitrosourea, anti-metabolites, such as methotrexate and hydroxyurea, and other agents, such as etoposides, campathecins, bleomycin, doxorubicin, daunorubicin, etc., although not necessarily cell cycle specific, kill cells during S phase because of their effect on DNA replication.
  • agents specifically colchicine and the vinca alkaloids, such as vinblastine and vincristine, interfere with microtubule assembly resulting in mitotic arrest.
  • Chemotherapy protocols generally involve administration of a combination of chemotherapeutic agents to increase the efficacy of treatment.
  • chemotherapeutic agents have many drawbacks ⁇ See, for example, Stockdale, 1998, "Principles Of Cancer Patient Management” in Scientific American Medicine, vol. 3, Rubenstein and Federman, eds., ch. 12, sect. 10). Almost all chemotherapeutic agents are toxic, and chemotherapy causes significant, and often dangerous, side effects, including severe nausea, bone marrow depression, immunosuppression, etc. Additionally, even with administration of combinations of chemotherapeutic agents, many tumor cells are resistant or develop resistance to the chemotherapeutic agents.
  • those cells resistant to the particular chemotherapeutic agents used in the treatment protocol often prove to be resistant to other drugs, even those agents that act by mechanisms different from the mechanisms of action of the drugs used in the specific treatment; this phenomenon is termed pleiotropic drug or multidrug resistance.
  • drug resistance many cancers prove refractory to standard chemotherapeutic treatment protocols.
  • Immune-mediated allergic (hypersensitivity) reactions are classified into four types (MV) according to the underlying mechanisms leading to the expression of the allergic symptoms.
  • Type I allergic reactions are characterized by IgE-mediated release of vasoactive substances such as histamine from mast cells and basophils. The release of these substances and the subsequent manifestation of allergic symptoms are initiated by the cross-linking of allergen- bound IgE to its receptor on the surface of mast cells and basophils.
  • IgE-mediated release of vasoactive substances such as histamine from mast cells and basophils.
  • the release of these substances and the subsequent manifestation of allergic symptoms are initiated by the cross-linking of allergen- bound IgE to its receptor on the surface of mast cells and basophils.
  • IgE antibodies specific for the allergen in individuals suffering from type I allergic reactions, exposure to an allergen for a second time leads to the production of high levels of IgE antibodies specific for the allergen as a result of the involvement of memory B and T cells in the 3 -cell interaction required for IgE
  • IgE antibodies produced cause an increase in the cross-linking of IgE receptors on mast cells and basophils by allergen-bound IgE, which in turn leads to the activation of these cells and the release of the pharmacological mediators that are responsible for the clinical manifestations of type I allergic diseases.
  • the high affinity receptor (Fc ⁇ RI) is expressed on the surface of mast cells and basophils.
  • the low affinity receptor (Fc ⁇ RIl/CD23) is expressed on many cell types including B cells, T cells, macrophages, eosinophils and Langerhan cells.
  • the high affinity IgE receptor consists of three subunits (alpha, beta and gamma chains).
  • NYJD 1603354 2 allergies For example, the elucidation of the IgE receptor-binding site could lead to the identification of peptides or small molecules that block the binding of IgE to receptor-bearing cells in vivo.
  • IgE-mediated allergic reactions are treated with drugs such as antihistamines and corticosteroids which attempt to alleviate the symptoms associated with allergic reactions by counteracting the effects of the vasoactive substances released from mast cells and basophils.
  • drugs such as antihistamines and corticosteroids which attempt to alleviate the symptoms associated with allergic reactions by counteracting the effects of the vasoactive substances released from mast cells and basophils.
  • High doses of antihistamines and corticosteroids have deleterious side effects (e.g., central nervous system disturbance, constipation, etc).
  • other methods for treating type I allergic reactions are needed.
  • One approach to the treatment of type I allergic disorders has been the production of monoclonal antibodies which react with soluble (free) IgE in serum, block IgE from binding to its receptor on mast cells and basophils, and do not bind to receptor-bound IgE (i.e., they are non-anaphylactogenic).
  • Two such monoclonal antibodies are in advanced stages of clinical development for treatment of IgE-mediated allergic reactions (see, e.g., Chang, T.W., 2000, Nature Biotechnology 18:157-62).
  • NYJD 1603354.2 libraries with the monoclonal antibody BSWI7 to identify peptide mimotopes that could mimic the conformation of the IgE receptor binding. These mimotopes could presumably be used to induce polyclonal antibodies that react with free native IgE, but not with receptor-bound IgE as well as block IgE from binding to its receptor.
  • Kriek et al. disclosed peptide mimotopes that are not homologous to any part of the IgE molecule and are thus different from peptides disclosed in the present invention.
  • Fc ⁇ RIIA and Fc ⁇ RIIB exhibit very different activities.
  • the fundamental difference is that the Fc ⁇ RIIA initiates intracellular signaling leading to cell activation such as phagocytosis and respiratory burst, whereas the Fc ⁇ RIIB initiates inhibitory signaling.
  • antibodies known to distinguish among native human Fc ⁇ RIIA and native human Fc ⁇ RIIB have not been identified; in view of their distinctive activities and role in modulating immune responses, such antibodies that recognize native Fc ⁇ RIIB, and not native Fc ⁇ RIIA, are needed.
  • the present invention is based, in part, on the discovery of such Fc ⁇ RIIB -specific antibodies.
  • “native Fc ⁇ RIIB or Fc ⁇ RIIA” means Fc ⁇ RIIB or Fc ⁇ RIIA which is endogenously expressed in a cell and is present on the cell surface of that cell or recombinantly expressed in a mammalian cell and present on the cell surface, but is not Fc ⁇ RIIB or Fc ⁇ RIIA expressed in a bacterial cell or denatured, isolated Fc ⁇ RIIB or Fc ⁇ RIIA.
  • the invention relates to an antibody or a fragment thereof that specifically binds
  • Fc ⁇ RIIB particularly human Fc ⁇ RIIB, more particularly native human Fc ⁇ RIIB
  • the antibodies of the invention bind the extracellular domain of native human Fc ⁇ RIIB.
  • the antibody or a fragment thereof binds Fc ⁇ RIIB with at least 2 times greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA.
  • the antibody or a fragment thereof binds
  • NYJD 1603354.2 Fc ⁇ RIIB with at least 4 times, at least 6 times, at least 8 times, at least 10 times, at least 100 times, at least 1000 times, at least 10 4 , at least 10 5 , at least 10 6 , at least 10 7 , or at least 10 8 times greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA
  • said antibody or a fragment thereof binds Fc ⁇ RIIB with 100 times, 1000 times, 10 4 times , 10 5 times, 10 6 times, 10 7 times, or 10 8 times greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA.
  • these binding affinities are determined with the monomeric IgG, and not the aggregated IgG, and binding is via the variable domain (e.g., Fab fragments of the antibodies have binding characteristic similar to the full immunolobulin molecule).
  • the anti-Fc ⁇ RIIB antibodies block the ligand binding site of Fc ⁇ RIIB.
  • the blocking activity can block the negative regulation of immune-complex-triggered activation and consequently enhance the immune response.
  • the enhanced immune response is an increase in antibody-dependent cellular response.
  • the anti- Fc ⁇ RIIB antibodies of the invention block crosslinking of Fc ⁇ RIIB receptors to B cell and/or Fc receptors, leading to B cell, mast cell, dendritic cell, or macrophage activation.
  • the antibody or fragment thereof blocks crosslinking of Fc ⁇ RIIB to an immunoreceptor tyrosine-based activation motif (ITAM) containing activating receptor, preferably enhancing the activity of an activating receptor.
  • ITAM-containing recpetors include Fc receptors, and BCR-associated Ig ⁇ .
  • the blocking leads to B cell, mast cell, dendritic cell, or macrophage activation.
  • the Fc receptor is a Fc ⁇ R or a Fc ⁇ R, preferably Fc ⁇ RI.
  • an Fc ⁇ RI dependent activity is modulated, for example, modulation of calcium mobilization and/or modulation of degranulation.
  • the Fc ⁇ RIIB -specific antibody in accordance with the invention is not the monoclonal antibody designated KB61, as disclosed in Pulford et al., 1986 (Immunology, 57: 71-76) or the monoclonal antibody designated MAbII8D2 as disclosed in Weinrich et al., 1996, (Hybridoma, 15(2):109-6).
  • the Fc ⁇ RIIB- specific antibody of the invention does not bind to the same epitope and/or does not compete for binding with the monoclonal antibody KB61 or the monoclonal antibody MAbII8D2.
  • the Fc ⁇ RIIB-specific antibody of the invention does not bind the amino acid sequence Ser-Asp-Pro-Asn-Phe-Ser-Ile corresponding to amino acid positions 135-141 of Fc ⁇ RIIb2 isoform.
  • the invention relates to an isolated antibody or a fragment thereof that specifically binds Fc ⁇ RIIB with a greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA, and the constant domain of said antibody further has an enhanced affinity for at least one or more Fc activation receptors.
  • said Fc activation receptor is Fc ⁇ RIII.
  • said antibody or a fragment thereof blocks the IgG binding site of Fc ⁇ RIIB and blocks the binding of aggregated labeled IgGs to Fc ⁇ RIIB in, for example, a blocking ELISA assay.
  • said antibody or a fragment thereof blocks the binding of aggregated labeled IgGs in an ELISA blocking assay by at least 50%, 60%, 70%, 80%, 90%, 95%, 99%, or 99.9%.
  • the antibody or a fragment thereof completely blocks the binding of said aggregated labeled IgG in said ELISA assay.
  • said antibody or a fragment thereof blocks the IgG binding site of Fc ⁇ RIIB and blocks the binding of aggregated labeled IgG to
  • Fc ⁇ RIIB as determined by a double-staining FACS assay.
  • the invention encompasses the use of antibodies that modulate (i.e., agonize or antagonize) the activity of Fc ⁇ RIIB.
  • the antibodies of the invention agonize at least one activity of Fc ⁇ RIIB, i.e., elicit signaling.
  • agonistic antibodies of the invention may mimic clustering of Fc ⁇ RIIB leading to dampening of the activating response to Fc ⁇ R ligation and inhibition of cellular responsiveness.
  • the antibodies of the invention antagonize at least one activity of Fc ⁇ RIIB, i.e., block signaling.
  • the antibodies of the invention block the binding of aggregated IgGs to Fc ⁇ RIIB.
  • the invention provides antibodies that inhibit Fc ⁇ RI-induced mast cell activation.
  • the invention further provides anti-Fc ⁇ RIIB antibodies that inhibit Fc ⁇ RIIA-mediated macrophage activation in monocytic cells.
  • the invention also provides anti-Fc ⁇ RIIB antibodies that inhibit B-cell receptor mediated signaling.
  • the Fc region comprises at least one amino acid modification relative to a wild-type Fc region, such that the modified Fc region has an altered binding affinity to a Fc receptor.
  • the antibody or fragment thereof has an increased binding affinity to Fc ⁇ RIIB or Fc ⁇ RIII.
  • Preferred amino acid modifications comprise a substitution at position 265 or 297. More preferably, the amino acid modification is a substitution at position 265 with alanine or a substitution at position 297 with glutamine.
  • the invention provides a monoclonal antibody produced by clone 2B6 or 3H7, having ATCC accession numbers PTA-4591 and PTA-4592, respectively.
  • the invention provides an isolated antibody or a fragment thereof that competes for binding with the monoclonal antibody produced by clone 2B6 or 3H7 and binds Fc ⁇ RIIB, preferably native human Fc ⁇ RIIB with a greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA, preferably native human Fc ⁇ RIIA and/or binds to the same epitope of Fc ⁇ RIIB as the monoclonal antibody produced from clone 2B6 or 3H7 and binds Fc ⁇ RIIB with a greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA.
  • the invention provides hybridoma cell line 2B6 or 3H7, having ATCC accession numbers PTA
  • the methods of the invention also encompass polynucleotides that encode the antibodies of the invention.
  • the invention provides an isolated nucleic acid sequence encoding a heavy chain or a light chain of an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB with greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA.
  • the invention provides an isolated nucleic acid sequence encoding a heavy chain or a light chain of an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB and blocks the Fc binding domain of Fc ⁇ RIIB.
  • the invention also relates to a vector comprising said nucleic acid.
  • the invention further provides a vector comprising a first nucleic acid molecule encoding a heavy chain and a second nucleic acid molecule encoding a light chain, said heavy chain and light chain being of an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB with greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA.
  • the invention further provides a vector comprising a first nucleic acid molecule encoding a heavy chain and a second nucleic acid molecule encoding a light chain, said heavy chain and light chain being of an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB and blocks the Fc binding domain of Fc ⁇ RIIB.
  • said vector is an expression vector.
  • the invention further provides host cells containing the vectors of or polynucleotides encoding the antibodies of the invention.
  • the invention encompasses polynucleotides encoding heavy and light chains of the antibodies produced by the deposited hybridoma clones, having ATCC accession numbers PTA-4591 and PTA-4592, respectively, or portions thereof, e.g., CDRs, variable domains, etc. and humanized versions thereof.
  • the invention further provides methods for the production of antibodies of the invention or fragments thereof.
  • the antibodies of the invention or fragments thereof can be produced by any method known in the art for the production of antibodies, in particular, by secretion from cultured hybridoma cells, chemical synthesis or by recombinant expression
  • the invention relates to a method for recombinantly producing a Fc ⁇ RIIB- specific antibody, said method comprising: (i) culturing under conditions suitable for the expression of said antibody in a medium, a host cell containing a first nucleic acid molecule, operably linked to a heterologous promoter and a second nucleic acid operably linked to the same or a different heterologous promoter, said first nucleic acid and second nucleic acid encoding a heavy chain and a light chain, respectively, of an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB with greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA or an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB and blocks the Fc binding domain of Fc ⁇ RIIB; and (ii) recovery of said antibody from said medium.
  • the antibodies of the invention are monoclonal antibodies, and more preferably, humanized or human antibodies.
  • an antibody fragment of the invention is a F(ab') 2 fragment or F(ab) fragment.
  • the antibodies of the invention bind to the extracellular domain of human Fc ⁇ RIIB, particularly native human Fc ⁇ RIIB.
  • the antibodies of the invention specifically or selectively recognize one or more epitopes of Fc ⁇ RIIB, particularly native human Fc ⁇ RIIB.
  • Another embodiment of the invention encompasses the use of phage display technology to increase the affinity of the antibodies of the invention for Fc ⁇ RIIB.
  • antibodies of the invention are screened using antibody screening assays well known in the art (e.g., BIACORE assays) to identify antibodies with Koff rate less than 3x10 3 s "1 .
  • Activating and inhibitory Fc receptors are critical for the balanced function of these receptors and proper cellular immune responses.
  • the invention encompasses the use of the antibodies of the invention for the treatment of any disease related to loss of such balance and regulated control in the Fc receptor signaling pathway.
  • the Fc ⁇ RIIB antibodies of the invention have uses in regulating the immune response.
  • the Fc ⁇ RIIB antibodies of the invention can also be used to alter certain effector functions to enhance, for example, therapeutic antibody-mediated cytotoxicity.
  • the antibodies of the invention are useful for prevention or treatment of cancer, for example, in one embodiment, as a single agent therapy.
  • the antibodies of the invention are useful for prevention or treatment of B-cell malignancies, particularly non-Hodgkin's lymphoma or chronic lymphocytic leukemia.
  • the antibodies of the invention are used for the treatment and/or prevention of
  • the antibodies are useful for prevention or treatment of cancer, particularly in potentiating the cytotoxic activity of cancer antigen-specific therapeutic antibodies with cytotoxic activity to enhance tumor cell killing and/or enhancing antibody dependent cytotoxic cellular ("ADCC”) activity, complement dependent cytotoxic (“CDC”) activity, or phagocytosis of the therapeutic antibodies.
  • ADCC antibody dependent cytotoxic cellular
  • CDC complement dependent cytotoxic
  • the invention provides a method of treating cancer in a patient having a cancer characterized by a cancer antigen, said method comprising administering to said patient a therapeutically effective amount of a first antibody or a fragment thereof that specifically binds Fc ⁇ RIIB with greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA, and a second antibody that specifically binds said cancer antigen and is cytotoxic.
  • the invention also provides a method of treating cancer in a patient having a cancer characterized by a cancer antigen, said method comprising administering to said patient a therapeutically effective amount of an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB, particularly native human Fc ⁇ RIIB with greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA, preferably native human Fc ⁇ RIIA, and the constant domain of which further has an increased affinity for one or more Fc activation receptors, when the antibody is monomeric, such as Fc ⁇ RIIIA, and an antibody that specifically binds said cancer antigen and is cytotoxic.
  • said Fc activation receptor is Fc ⁇ RIIIA.
  • the invention also provides a method of treating cancer in a patient having a cancer characterized by a cancer antigen, said method comprising administering to said patient a therapeutically effective amount of an antibody or a fragment thereof that specifically binds said cancer antigen and a therapeutically effective amount of an antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB.
  • the invention provides a method of enhancing an antibody mediated cytotoxic effect in a subject being treated with a cytotoxic antibody, said method comprising administering to said patient an antibody of the invention or a fragment thereof, in an amount sufficient to enhance the cytotoxic effect of said cytotoxic antibody.
  • the invention provides a method of enhancing an antibody-mediated cytotoxic effect in a subject being treated with a cytotoxic antibody, said method comprising administering to said patient an antibody of the invention or a fragment thereof, further having an enhanced affinity for an Fc activation receptor, when monomeric, in an amount sufficient to enhance the cytotoxic effect of said cytotoxic antibody.
  • the invention provides a method further comprising the administration of one or more additional cancer therapies.
  • the invention provides a method of regulating immune-complex mediated cell activation in a patient, said method comprising administering to said patient a therapeutically effective amount of an antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB.
  • administration of the antibody or fragment thereof results in an enhanced immune response, such as an increase in an antibody-dependent cellular response.
  • the immune complex mediated cell activation is B cell activation, mast cell activation, dendritic cell activation or macrophage activation.
  • the invention provides a method of breaking tolerance to an antigen in a patient, said method comprising administering to a patient in need thereof (1) an antigen-antibody complex comprising said antigen and (2) an antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB, thereby breaking tolerance in said patient to said antigen.
  • the antibody or fragment thereof can be administered before, concurrently with, or after administration of said antigen-antibody complex.
  • the invention futher provides a pharmaceutical composition
  • a pharmaceutical composition comprising (i) a therapeutically effective amount of an antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB; (ii) a cytotoxic antibody that specifically binds a cancer antigen; and (iii) a pharmaceutically acceptable carrier.
  • the antibody or fragment thereof is a human or humanized antibody.
  • the antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB blocks crosslinking of Fc ⁇ RIIB to a Fc receptor.
  • the antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB comprises a Fc region comprising at least one amino acid modification relative to a wild-type Fc region, such that the modified Fc region has an altered binding affinity to a Fc receptor.
  • the amino acid modification comprises a substitution at position 265 or 297, preferably a substitution at position 265 with alanine or a substitution at position 297 with glutamine.
  • the cytotoxic antibody is Herceptin®, Rituxan®, IC 14, PANOREXTM, IMC-225, VITAXINTM, Campath 1H/LDP-03, LYMPHOCIDETM, or ZEVLINTM.
  • the invention encompasses the use of the antibodies of the invention in combination with any therapeutic antibody that mediates its therapeutic effect through cell
  • the antibodies of the invention potentiate the antibody's therapeutic activity by enhancing antibody- mediated effector function.
  • the antibodies of the invention potentiate the cytotoxic antibody's therapeutic activity by enhancing phagocytosis and opsonization of the targeted tumor cells.
  • the antibodies of the invention potentiate the antibody's therapeutic activity by enhancing antibody- dependent cell-mediated cytotoxicity ("ADCC") in destruction of the targeted tumor cells.
  • ADCC antibody- dependent cell-mediated cytotoxicity
  • the invention encompasses use of the antibodies of the invention in combination with a therapeutic antibody that does not mediate its therapeutic effect through cell killing to potentiate the antibody's therapeutic activity.
  • the invention encompasses use of the antibodies of the invention in combination with a therapeutic apoptosis inducing antibody with agonistic activity, e.g., anti-Fas antibody.
  • Therapeutic apoptosis inducing antibodies may be specific for any death receptor known in the art for the modulation of apoptotic pathway, e.g., TNFR receptor family member.
  • the invention encompasses using the antibodies of the invention to block macrophage mediated tumor cell progression and metastasis.
  • the antibodies of the invention are particularly useful in the treatment of solid tumors, where macrophage infiltration occurs.
  • the antagonistic antibodies of the invention are particularly useful for controlling, e.g., reducing or eliminating, tumor cell metastasis, by reducing or eliminating the population of macrophages that are localized at the tumor site.
  • the invention further encompasses antibodies that effectively deplete or eliminate immune effector cells other than macrophages that express Fc ⁇ RIIB, e.g., dendritic cells.
  • Effective depletion or elimination of immune effector cells using the antibodies of the invention may range from a reduction in population of the effector cells by 50%, 60%, 70%, 80%, preferably 90%, and most preferably 99%.
  • the agonistic antibodies of the invention are particularly useful for the treatment of tumors of non-hematopoietic origin, including tumors of melanoma cells.
  • the term "specifically binds to Fc ⁇ RIIB" and analogous terms refer to antibodies or fragments thereof that specifically bind to Fc ⁇ RIIB or a fragment thereof and do not specifically bind to other Fc receptors, in particular to Fc ⁇ RIIA. Further it is understood to one skilled in the art, that an antibody that specifically binds to Fc ⁇ RIIB, may bind through the variable domain or the constant domain of the antibody. If the antibody that
  • NYJD: 1603354.2 specifically binds to Fc ⁇ RIIB binds through its variable domain, it is understood to one skilled in the art that it is not aggregated, i.e., is monomeric.
  • An antibody that specifically binds to Fc ⁇ RIIB may bind to other peptides or polypeptides with lower affinity as determined by, e.g., immunoassays, BIAcore, or other assays known in the art.
  • antibodies or fragments that specifically bind to Fc ⁇ RIIB or a fragment thereof do not cross-react with other antigens.
  • Antibodies or fragments that specifically bind to Fc ⁇ RIIB can be identified, for example, by immunoassays, BIAcore, or other techniques known to those of skill in the art.
  • An antibody or a fragment thereof binds specifically to a Fc ⁇ RIIB when it binds to Fc ⁇ RIIB with higher affinity than to any cross-reactive antigen as determined using experimental techniques, such as western blots, radioimmunoassays (RIA) and enzyme-linked immunosorbent assays (ELISAs).
  • RIA radioimmunoassays
  • ELISAs enzyme-linked immunosorbent assays
  • the term “native Fc ⁇ RIIB” refers to Fc ⁇ RIIB which is endogenously expressed and present on the surface of a cell.
  • “native Fc ⁇ RIIB” encompasses a protein that is recombinantly expressed in a mammalian cell.
  • the native Fc ⁇ RIIB is not expressed in a bacterial cell, i.e., E. coli.
  • the native Fc ⁇ RIIB is not denatured, i.e., it is in its biologically active conformation.
  • the term “native Fc ⁇ RIIA” refers to Fc ⁇ RIIA which is endogenously expressed and present on the surface of a cell, hi some embodiments, "native Fc ⁇ RIIA” encompasses a protein that is recombinantly expressed in a mammalian cell.
  • the native Fc ⁇ RIIA is not expressed in a bacterial cell, i.e., E. coli. Most preferably the native Fc ⁇ RIIA is not denatured, i.e., it is in its biologically active conformation.
  • antibody refers to monoclonal antibodies, multispecific antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, camelized antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), intrabodies, and anti- idiotypic (anti-Id) antibodies (including, e.g., anti-Id and anti-anti-Id antibodies to antibodies of the invention), and epitope-binding fragments of any of the above.
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an antigen binding site.
  • Immunoglobulin molecules can be of any type ⁇ e.g., IgG, IgE, IgM, IgD, IgA and IgY), class ⁇ e.g., IgGj, IgG 2 , IgG 3 , IgG 4 , IgA 1 and IgA 2 ) or subclass.
  • derivative in the context of polypeptides or proteins refers to a polypeptide or protein that comprises an amino acid sequence which has been altered
  • NYJD 1603354.2 by the introduction of amino acid residue substitutions, deletions or additions.
  • derivative also refers to a polypeptide or protein which has been modified, Le, by the covalent attachment of any type of molecule to the polypeptide or protein.
  • an antibody may be modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc.
  • a derivative polypeptide or protein may be produced by chemical modifications using techniques known to those of skill in the art, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Further, a derivative polypeptide or protein derivative possesses a similar or identical function as the polypeptide or protein from which it was derived.
  • derivative refers to a polypeptide that comprises an amino acid sequence of a Fc ⁇ RIIB polypeptide, a fragment of a Fc ⁇ RIIB polypeptide, an antibody that immunospecifically binds to a Fc)RIIB polypeptide, or an antibody fragment that immunospecifically binds to a Fc)RIIB polypeptide, that has been altered by the introduction of amino acid residue substitutions, deletions or additions (i.e., mutations).
  • an antibody derivative or fragment thereof comprises amino acid residue substitutions, deletions or additions in one or more CDRs.
  • the antibody derivative may have substantially the same binding, better binding, or worse binding when compared to a non-derivative antibody, hi specific embodiments, one, two, three, four, or five amino acid residues of the CDR have been substituted, deleted or added (i.e., mutated).
  • derivative as used herein also refers to a Fc)RIIB polypeptide, a fragment of a Fc ⁇ RIIB polypeptide, an antibody that immunospecifically binds to a Fc)RIIB polypeptide, or an antibody fragment that immunospecifically binds to a Fc)RIIB polypeptide which has been modified, i.e., by the covalent attachment of any type of molecule to the polypeptide.
  • a Fc)RIIB polypeptide, a fragment of a Fc ⁇ RIIB polypeptide, an antibody, or antibody fragment may be modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc.
  • a derivative of a Fc ⁇ RIIB polypeptide, a fragment of a Fc ⁇ RIIB polypeptide, an antibody, or antibody fragment may be modified by chemical modifications using techniques known to those of skill in the art, including, but not limited to, specific chemical cleavage, acetylation, formulation, metabolic synthesis of tunicamycin, etc. Further, a derivative of a Fc ⁇ RIIB polypeptide, a fragment of a Fc ⁇ RIIB polypeptide, an antibody, or antibody fragment may contain one or more non-classical
  • a polypeptide derivative possesses a similar or identical function as a Fc)RIIB polypeptide, a fragment of a Fc)RIIB polypeptide, an antibody, or antibody fragment described herein.
  • a derivative of a Fc ⁇ RIIB polypeptide, a fragment of a Fc)RIIB polypeptide, an antibody, or antibody fragment has an altered activity when compared to an unaltered polypeptide.
  • a derivative antibody or fragment thereof can bind to its epitope more tightly or be more resistant to proteolysis.
  • autoimmune disease is used interchangeably with the term “autoimmune disorder” to refer to a condition in a subject characterized by cellular, tissue and/or organ injury caused by an immunologic reaction of the subject to its own cells, tissues and/or organs.
  • inflammatory disease is used interchangeably with the term “inflammatory disorder” to refer to a condition in a subject characterized by inflammation, preferably chronic inflammation.
  • Autoimmune disorders may or may not be associated with inflammation.
  • inflammation may or may not be caused by an autoimmune disorder.
  • certain disorders may be characterized as both autoimmune and inflammatory disorders.
  • cancer refers to a neoplasm or tumor resulting from abnormal uncontrolled growth of cells.
  • cancer explicitly includes, leukemias and lymphomas.
  • cancer refers to a disease involving cells that have the potential to metastasize to distal sites and exhibit phenotypic traits that differ from those of non-cancer cells, for example, formation of colonies in a three-dimensional substrate such as soft agar or the formation of tubular networks or weblike matrices in a three-dimensional basement membrane or extracellular matrix preparation.
  • Non-cancer cells do not form colonies in soft agar and form distinct sphere-like structures in three-dimensional basement membrane or extracellular matrix preparations.
  • Cancer cells acquire a characteristic set of functional capabilities during their development, albeit through various mechanisms. Such capabilities include evading apoptosis, self-sufficiency in growth signals, insensitivity to anti-growth signals, tissue invasion/metastasis, limitless explicative potential, and sustained angiogenesis.
  • the term "cancer cell” is meant to encompass both pre-malignant and malignant cancer cells.
  • cancer refers to a benign tumor, which has remained localized.
  • cancer refers to a malignant tumor, which has invaded and destroyed neighboring body structures and spread to distant sites.
  • the cancer is associated with a specific cancer antigen.
  • immunomodulatory agent refers to an agent that modulates a host's immune system.
  • an immunomodulatory agent is an immunosuppressant agent.
  • an immunomodulatory agent is an immunostimulatory agent.
  • Immunomodatory agents include, but are not limited to, small molecules, peptides, polypeptides, fusion proteins, antibodies, inorganic molecules, mimetic agents, and organic molecules.
  • epitope refers to a fragment of a polypeptide or protein having antigenic or immunogenic activity in an animal, preferably in a mammal, and most preferably in a human.
  • An epitope having immunogenic activity is a fragment of a polypeptide or protein that elicits an antibody response in an animal.
  • An epitope having antigenic activity is a fragment of a polypeptide or protein to which an antibody immunospecifically binds as determined by any method well-known to one of skill in the art, for example by immunoassays. Antigenic epitopes need not necessarily be immunogenic.
  • fragment refers to a peptide or polypeptide comprising an amino acid sequence of at least 5 contiguous amino acid residues, at least 10 contiguous amino acid residues, at least 15 contiguous amino acid residues, at least 20 contiguous amino acid residues, at least 25 contiguous amino acid residues, at least 40 contiguous amino acid residues, at least 50 contiguous amino acid residues, at least 60 contiguous amino residues, at least 70 contiguous amino acid residues, at least contiguous 80 amino acid residues, at least contiguous 90 amino acid residues, at least contiguous 100 amino acid residues, at least contiguous 125 amino acid residues, at least 150 contiguous amino acid residues, at least contiguous 175 amino acid residues, at least contiguous 200 amino acid residues, or at least contiguous 250 amino acid residues of the amino acid sequence of another polypeptide.
  • a fragment of a polypeptide retains at least one function of the polypeptide.
  • antibody fragments are epitope binding fragments.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which hypervariable region residues of the recipient are replaced by hypervariable region residues from a non-human species (donor antibody) such as mouse, rat, rabbit or non-human primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or non-human primate having the desired specificity, affinity, and capacity.
  • framework Region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non-human immunoglobulin and all or substantially all of the P 7 Rs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin that immunospecifically binds to a FcyRIIB polypeptide, that has been altered by the introduction of amino acid residue substitutions, deletions or additions (i.e., mutations).
  • Fc immunoglobulin constant region
  • a humanized antibody is a derivative.
  • Such a humanized antibody comprises amino acid residue substitutions, deletions or additions in one or more non-human CDRs.
  • the humanized antibody derivative may have substantially the same binding, better binding, or worse binding when compared to a non- derivative humanized antibody.
  • one, two, three, four, or five amino acid residues of the CDR have been substituted, deleted or added (i.e., mutated).
  • European Patent Nos. EP 239,400, EP 592,106, and EP 519,596 International Publication Nos. WO 91/09967 and WO 93/17105; U.S. Patent Nos.
  • hypervariable region refers to the amino acid residues of an antibody which are responsible for antigen binding.
  • the hypervariable region comprises amino acid residues from a "Complementarity Determining Region” or "CDR" (i.e., residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (Hl), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD.
  • CDR Constantarity Determining Region
  • residues from a "hypervariable loop” i.e., residues 26-32 (Ll), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (Hl), 53-55 (H2) and 96-101
  • single-chain Fv or “scFv” refer to antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding.
  • scFvs include bi-specific scFvs and humanized scFvs.
  • DNA molecules ⁇ e.g., cDNA or genomic DNA
  • RNA molecules ⁇ e.g., mRNA
  • combinations of DNA and RNA molecules or hybrid DNA/RNA molecules and analogs of DNA or RNA molecules.
  • analogs can be generated using, for example, nucleotide analogs, which include, but are not limited to, inosine or tritylated bases.
  • Such analogs can also comprise DNA or RNA molecules comprising modified backbones that lend beneficial attributes to the molecules such as, for example, nuclease resistance or an increased ability to cross cellular membranes.
  • nucleic acids or nucleotide sequences can be single-stranded, double-stranded, may contain both single-stranded and double-stranded portions, and may contain triple-stranded portions, but preferably is double-stranded DNA.
  • a subject is preferably a mammal such as a non-primate ⁇ e.g., cows, pigs, horses, cats, dogs, rats etc.) and a primate ⁇ e.g., monkey and human), most preferably a human.
  • a primate e.g., monkey and human
  • the terms “treat,” “treating” and “treatment” refer to the eradication, reduction or amelioration of symptoms of a disease or disorder related to the loss of regulation in the Fc receptor signaling pathway or to enhance the therapeutic efficacy of another therapy, e.g., a therapeutic antibody, vaccine therapy.
  • treatment refers to the eradication, removal, modification, or control of primary, regional, or metastatic cancer tissue that results from the administration of one or more therapeutic agents.
  • such terms refer to the minimizing or delaying the spread of cancer resulting from the administration of one or more therapeutic agents to a subject with such a disease.
  • side effects encompasses unwanted and adverse effects of a prophylactic or therapeutic agent. Adverse effects are always unwanted, but
  • NYJD: 1603354.2 unwanted effects are not necessarily adverse.
  • An adverse effect from a prophylactic or therapeutic agent might be harmful or uncomfortable or risky.
  • Side effects from chemotherapy include, but are not limited to, gastrointestinal toxicity such as, but not limited to, early and late-forming diarrhea and flatulence, nausea, vomiting, anorexia, leukopenia, anemia, neutropenia, asthenia, abdominal cramping, fever, pain, loss of body weight, dehydration, alopecia, dyspnea, insomnia, dizziness, mucositis, xerostomia, and kidney failure, as well as constipation, nerve and muscle effects, temporary or permanent damage to kidneys and bladder, flu-like symptoms, fluid retention, and temporary or permanent infertility.
  • Side effects from radiation therapy include but are not limited to fatigue, dry mouth, and loss of appetite.
  • Side effects from biological therapies/immunotherapies include but are not limited to rashes or swellings at the site of administration, flu-like symptoms such as fever, chills and fatigue, digestive tract problems and allergic reactions.
  • Side effects from hormonal therapies include but are not limited to nausea, fertility problems, depression, loss of appetite, eye problems, headache, and weight fluctuation. Additional undesired effects typically experienced by patients are numerous and known in the art, see, e.g., the Physicians' Desk Reference (56 th ed., 2002), which is incorporated herein by reference in its entirety.
  • a "therapeutically effective amount” refers to that amount of the therapeutic agent sufficient to treat or manage a disease or disorder associated with Fc ⁇ RIIB and any disease related to the loss of regulation in the Fc receptor signaling pathway or to enhance the therapeutic efficacy of another therapy, e.g., therapeutic antibody, vaccine therapy, etc.
  • a therapeutically effective amount may refer to the amount of therapeutic agent sufficient to delay or minimize the onset of disease, e.g., delay or minimize the spread of cancer.
  • a therapeutically effective amount may also refer to the amount of the therapeutic agent that provides a therapeutic benefit in the treatment or management of a disease.
  • a therapeutically effective amount with respect to a therapeutic agent of the invention means that amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of a disease, e.g., sufficient to enhance the therapeutic efficacy of a therapeutic antibody sufficient to treat or manage a disease.
  • the term can encompass an amount that improves overall therapy, reduces or avoids unwanted effects, or enhances the therapeutic efficacy of or synergies with another therapeutic agent.
  • prophylactic agent and “prophylactic agents” refer to any agent(s) which can be used in the prevention of a disorder, or prevention of recurrence or spread of a disorder.
  • a prophylactically effective amount may refer to the amount of
  • NYJD 16033542 prophylactic agent sufficient to prevent the recurrence or spread of hyperproliferative disease, particularly cancer, or the occurrence of such in a patient, including but not limited to those predisposed to hyperproliferative disease, for example those genetically predisposed to cancer or previously exposed to carcinogens.
  • a prophylactically effective amount may also refer to the amount of the prophylactic agent that provides a prophylactic benefit in the prevention of disease.
  • a prophylactically effective amount with respect to a prophylactic agent of the invention means that amount of prophylactic agent alone, or in combination with other agents, that provides a prophylactic benefit in the prevention of disease.
  • the term can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of or synergies with another prophylactic agent, such as but not limited to a therapeutic antibody.
  • the term “prophylactic agent” refers to an agonistic Fc)RIIB -specific antibody.
  • the term “prophylactic agent” refers to an antagonistic Fc ⁇ RIIB- specific antibody.
  • the term “prophylactic agent” refers to cancer chemotherapeutics, radiation therapy, hormonal therapy, biological therapy (e.g., immunotherapy), and/or Fc ⁇ RIIB antibodies of the invention.
  • more than one prophylactic agent may be administered in combination.
  • the terms “manage,” “managing” and “management” refer to the beneficial effects that a subject derives from administration of a prophylactic or therapeutic agent, which does not result in a cure of the disease.
  • a subject is administered one or more prophylactic or therapeutic agents to "manage” a disease so as to prevent the progression or worsening of the disease.
  • the terms “prevent”, “preventing” and “prevention” refer to the prevention of the recurrence or onset of one or more symptoms of a disorder in a subject resulting from the administration of a prophylactic or therapeutic agent.
  • the term “in combination” refers to the use of more than one prophylactic and/or therapeutic agents. The use of the term “in combination” does not restrict the order in which prophylactic and/or therapeutic agents are administered to a subject with a disorder, e.g., hyperproliferative cell disorder, especially cancer.
  • a first prophylactic or therapeutic agent can be administered prior to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45
  • NYJD 1603354.2 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second prophylactic or therapeutic agent to a subject which had, has, or is susceptible to a disorder.
  • the prophylactic or therapeutic agents are administered to a subject in a sequence and within a time interval such that the agent of the invention can act together with the other agent to provide an increased benefit than if they were administered otherwise. Any additional prophylactic or therapeutic agent can be administered in any order with the other additional prophylactic or therapeutic agents.
  • FIGS. 1 A and B Direct binding of the antibody produced from the 3H7 clone to Fc ⁇ RIIB and Fc ⁇ RIIA.
  • A The direct binding of antibodies from some of the hybridoma cultures to the Fc ⁇ RIIs were compared to a commercially available anti-Fc ⁇ RII antibody in an ELISA assay where the plate was coated with the receptors. Different dilutions (1:10) of the supernatants were incubated on the plate. The bound antibodies were detected with a goat anti- mouse HRP conjugated antibody and the absorbance was monitored at 650 nm.
  • B. The direct binding of the antibody from the 3H7 hybridoma culture (supernatant n. 7 from the FIG. IA), in crude (left panel) and purified form (right panel), to Fc ⁇ RIIA and Fc ⁇ RIIB, were compared using the same ELISA assay as in IA.
  • FIG. 2 Competition in binding to Fc ⁇ RIIB of the antibody produced from the
  • 3H7 hybridoma and aggregated biotinylated human IgG The ability of the 3H7 antibody to compete with aggregated biotinylated human IgG for binding to Fc ⁇ RIIB was measured using a blocking ELISA experiment.
  • the ELISA plate coated with Fc ⁇ RIIB was incubated with the supernatant containing the 3H7 antibody and with a supernatant from the same hybridoma cells but not containing antibody (negative control).
  • FIG. 3 Comparison of the direct binding of the 3H7 antibody to Fc ⁇ RIIB produced in a bacterial or in a mammalian system. Direct binding of the 3H7 antibody to Fc ⁇ RIIB was measured using an ELISA assay. Binding to the bacterial or mammalian produced Fc ⁇ RIIB was compared.
  • the antibody titration started from the straight supernatant followed by 1:10 dilutions.
  • the bound antibody was detected with a goat anti-mouse HRP conjugated antibody, the reaction was developed with TMB and the absorbance was monitored at 650 nm.
  • FIG. 4 Direct binding of the 3H7 antibody to Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIIA .
  • the direct binding of the purified 3H7 antibody to Fc ⁇ RIIA, Fc ⁇ RIIB and Fc ⁇ RIIIA expressed in a mammalian system were compared using the ELISA assay.
  • ELISA plate was coated with the three receptors (100 ng/well). Different dilutions of the purified 3H7 antibody were incubated on the coated plate. A goat anti-mouse-HRP conjugated antibody was used for detection of the bound specific antibody, the reaction was developed with TMB and the absorbance was monitored at 650 run.
  • FIG. 5 Comparison of the direct binding ability to Fc ⁇ RIIA and Fc ⁇ RIIB of the antibody purified from clone 2B6 compared to other three commercially available monoclonal antibodies against Fc ⁇ RII.
  • the binding of 2B6 antibody to Fc ⁇ RIIA (top right panel) and Fc ⁇ RIIB (top left panel) is compared to that of three other commercially available antibodies raised against Fc ⁇ RII.
  • the ELISA format used is the same described in FIG. 4.
  • FIGS. 6 A and B. Competition in binding of the antibody produced from clone
  • FIGS. 7 A-C Competition of 2B6 antibody and aggregated biotinylated human IgG
  • a double staining FACS assay was performed to characterize the 2B6 antibody using CHO-Kl cells that had been stably transfected with full-length mammalian Fc ⁇ RIIB.
  • B The transfectant cells were stained with aggregated biotinylated human IgG after being stained with mouse IgGl isotype control and labeled with a goat anti-mouse-FITC conjugated antibody to detect the bound monoclonal antibody and with Streptavidin-PE conjugated to detect the bound aggregates.
  • C The cells were stained with 2B6 antibody, the
  • NYJD 16033542 antibody was removed by washes and the cells were incubated with aggregated biotinylated human IgG. Cells were washed and labeled with a goat anti-mouse-FITC conjugated antibody to detect the bound monoclonal antibody and with Streptavidin-PE conjugated to detect the bound aggregates.
  • FIGS. 8A-C Monoclonal anti Fc ⁇ RIIB antibodies and CD20 co-stain of human
  • B lymphocytes Cells from human blood (“buffy coat") were stained with anti-CD20 -FITC conjugated antibody, to select the B lymphocytes population, as well as 3H7 and 2B6. The bound anti-Fc ⁇ RIIB antibodies were detected with a goat anti-mouse-PE conjugated antibody.
  • B. Cells were co-stained with anti-CD20-FITC antibody and 3H7 antibody.
  • FIGS. 9 A and B Staining of CHO cells expressing Fc ⁇ RIIB.
  • A. CHO/IIB cells were stained with mouse IgGl isotype control (left panel) and 3H7 antibody (right panel).
  • B. CHO/IIB cells were stained with mouse IgGl isotype control (left panel) and 2B6 antibody (right panel). The cell-bound antibodies were labeled with a goat anti-mouse-PE conjugated antibody.
  • FIG. 10 Staining of CHO cells expressing Fc ⁇ RIIB.
  • CHO cells expressing huFc ⁇ RIIB were incubated with the anti-CD32B antibodies, indicated on top of each panel.
  • Cells were washed and 9 ⁇ g/ml of aggregated human IgG were added to the cells on ice.
  • the human aggregated IgG were detected with goat anti-human-IgG FITC conjugated.
  • Samples were analyzed by FACS isotype control + goat anti hulgG-FITC, — isotype control + aggregated humanlgG + goat anti humanlgG-FITC, — anti-CD32B antibody + aggregated humanlgG + goat anti humanlgG-FITC.
  • the amount of each antibody bound to the receptor on the cells was also detected (inset) on a separate set of samples using a goat anti-mouse PE conjugated antibody.
  • FIG. 11 Staining of Human PBMCs with 2B6, 3H7 and IV.3 Antibodies.
  • Human PBMCs were stained with 2B6, 3H7, and IV.3 antibodies, as indicated in the right side of the panel, followed by a goat anti-mouse-Cyanine(Cy5) conjugated antibody; two color staining using anti-CD20-FITC conjugated for B lymphocytes, anti-CD 14-PE conjugated for monocytes, anti-CD56-PE conjugated for NK cells and anti-CD16-PE conjugated for granulocytes.
  • FIGS. 12 A and B ⁇ -Hexaminidase Release Assay.
  • A Schematic representation of ⁇ -hexaminidase release assay. Transfectants expressing human Fc ⁇ RIIB were sensitized with
  • Crosslinking occurs because of the ability of the polyclonal antibody to recognize the light chain of the murine IgE antibody bound to Fc ⁇ RI.
  • Transfectants sensitized with murine IgE and preincubated with 2B6 antibody were also challenged with F(ab') 2 fragments of a polyclonal goat anti-mouse IgG to cross link Fc ⁇ RI to FC7RIIB.
  • GAM F(ab) 2 goat anti-mouse F(ab) 2 fragment
  • ⁇ -hexosaminidase activity released in the supernatant and within the cells was determined by a colorimetric assay using p-nitrophenyl N-acetyl- ⁇ -D-glucosaminide.
  • the released ⁇ -hexosaminidase activity was expressed as a percentage of the released activity relative to the total activity.
  • FIGS. 13A-C. 2B6 is capable of functionally blocking the Fc binding site of
  • A Schematic representation of the experimental model.
  • B and C RBL-2H3/CD32B cells were stimulated with BSA-DNP-FITC complex in the presence of human IgGl, with BSA-DNP-FITC complexed with chimeric D265A4-4-20 in the presence or not of 3 ⁇ g/ml of F(ab)2 fragments of 2B6 (B).
  • Cells were also stimulated with BSA-DNP-FITC complex in the presence of human IgGl, with BSA-DNP-FITC complexed with chimeric 4-4-20 in the presence or not of 3 ⁇ g/ml of F(ab)2 fragments of 2B6 (C).
  • B-hexosaminidase activity released in the supernatant and within the cells was determined by a colorimetric assay using p- nitrophenyl N-acetyl- ⁇ -D-glucosaminide. The released ⁇ -hexosaminidase activity was expressed as a percentage of the released activity relative to the total activity.
  • FIGS. 14 A-C Ovarian and Breast carcinoma cell lines express Her2/neu to varying levels. Staining of A: Ovarian IGROV-I with purified ch4D5, B: Ovarian OVCAR-8 with purified 4D5 antibody, and C: Breast cancer SKBR-3 cells with purified ch4D5 followed by goat anti-human-conjugated to phycoerythrin (PE). The relevant isotype control IgGl is indicated the left of the staining with anti-Her2neu antibody.
  • PE phycoerythrin
  • FIGS. 15 A-C Elutriated Monocytes express all Fc ⁇ Rs: A. MDM obtained from donor 1; B. MDM obtained from donor 2; propagated in human serum or human serum and GMCSF; C. Monocytes thawed and stained immediately. Monocyte-derived
  • NYJD 1603354.2 macrophages were stained with anti-bodies specific for human Fc ⁇ R receptor.
  • the solid histogram in each plot represents the background staining.
  • the clear histogram within each panel represents the staining with specific anti-human Fc ⁇ R antibodies.
  • FIGS. 16A and B Ch4D5 mediates effective ADCC with ovarian and breast cancer cell lines using PBMC. Specific lysis subtracted from antibody-independent lysis is shown for A. Ovarian tumor cell line, IGROV-I at an effector: target ratio of 75: 1, and for B.
  • FIGS. 17A-C Histochemical staining of human ovarian ascites shows tumors cells and other inflammatory cells.
  • neoplastic cells can be identified by the irregular size and shape, scattered cytoplasm, and irregular dense nuclei.
  • B. Giemsa stain of unprocessed ascites from a patient with serous tumor of the ovary shows two mesothelial cells placed back to back indicated by short arrows. Also shown is a cluster of five malignant epithelial cells indicated by the long arrow. Erythrocytes are visible in the background.
  • FIG. 18 In vitro ADCC assay of ch2B6 and aglycosylated ch2B6 in Daudi cells. ch2B6 antibody mediates in vitro ADCC in CD32B expressing daudi cells.
  • FIG. 19 In vitro ADCC assay of ch 2B6 and aglycosylated ch2B6 in Raji cells. ch2B6 antibody mediates in vitro ADCC in CD32B expressing Raji cells.
  • FIG. 20 Estimated tumor size in individual mice. Injection days are indicated by arrows.
  • the invention encompasses antibodies (preferably monoclonal antibodies) or fragments thereof that specifically bind Fc ⁇ RIIB, preferably human Fc ⁇ RIIB, more preferably native human Fc ⁇ RIIB with a greater affinity than said antibodies or fragments thereof bind Fc ⁇ RIIA, preferably human Fc ⁇ RIIA, more preferably native human Fc ⁇ RIIA.
  • the antibodies of the invention bind the extracellular domain of native human Fc ⁇ RIIB.
  • the antibodies or fragments thereof bind to Fc ⁇ RIIB with an affinity greater than two-fold, four fold, 6 fold, 10 fold, 20 fold, 50 fold, 100 fold, 1000 fold, 10 4 fold, 10 5 fold , 10 6 fold, 10 7 fold, or 10 8 fold than said antibodies or fragments thereof bind Fc ⁇ RIIA.
  • the invention also encompasses antibodies or a fragments thereof that specifically binds Fc ⁇ RIIB, particularly human Fc ⁇ RIIB, more particularly native human Fc)RIIB, and blocks the Fc binding domain of Fc ⁇ RIIB, particularly human Fc ⁇ RIIB, more particularly native human Fc ⁇ RIIB.
  • the antibodies of the invention bind the extracellular domain of native human Fc ⁇ RIIB.
  • the antibody or fragment thereof blocks crosslinking of Fc ⁇ RIIB to an immunoreceptor tyrosine-based activation motif (ITAM) containing activating receptor.
  • ITAM immunoreceptor tyrosine-based activation motif
  • ITAM containing receptors include, but are not limited to Fc Receptors (CD64, CD32A, CD16, CD23, Fc ⁇ RI, etc.); TCR-associated CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , and ⁇ chains; BCR-associated Ig ⁇ (CD79A) and Ig ⁇ (CD79B) chains; DAP12; as well as several virally encoded transmembrane molecules. See Billadeau et ah, 2002, J. Clin. Invest. 109:161-168, herein incorporated by reference in its entirety. In preferred embodments, this blocking enhances the activity of the activating receptor and/or leads to B cell, mast cell, dendritic cell, or macrophage activation.
  • the Fc receptor is a Fc ⁇ R or a Fc ⁇ R, preferably Fc ⁇ RI.
  • an Fc ⁇ RI dependent activity is modulated.
  • the Fc ⁇ RI dependent activity is modulation of calcium mobilization and/or modulation of degranulation.
  • the antibody is a mouse monoclonal antibody produced by clone 2B6 or 3H7, having ATCC accession numbers PTA-4591 and PTA-4592, respectively.
  • Hybridomas producing antibodies of the invention have been deposited with the American Type Culture Collection (10801 University Boulevard., Manassas, VA. 20110-2209) on August 13, 2002 under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedures, and assigned accession numbers PTA-4591 (for hybridoma producing 2B6) and PTA-4592 (for hybridoma producing 3H7), respectively and are incorporated herein by reference.
  • the antibodies of the invention are human or have been humanized, preferably a humanized version of the antibody produced by clone 3H7 or 2B6.
  • the antibodies of the invention are human or have been humanized, preferably a humanized version of the antibody produced by clone 1D5, 2El, 2H9, 2Dl 1, or 1F2.
  • Humanized version of Fc ⁇ RIIB -specific antibodies are described in U.S. Application No. 11/126,978, filed May 10, 2005, herein incorporated by reference in its entirety.
  • the antibodies of the invention further do not bind Fc activation receptors, e.g., Fc ⁇ llLA, Fc ⁇ lllB, etc.
  • Fc ⁇ RIIB - specific antibody in accordance with the invention is not the monoclonal antibody designated
  • the Fc ⁇ RIIB -specific antibody of the invention does not bind to the same epitope and/or does not compete with binding with the monoclonal antibody KB61 or II8D2.
  • the Fc ⁇ RIIB -specific antibody of the invention does not bind the amino acid sequence SDPNFSI corresponding to positions 135-141 of Fc ⁇ RIIb2 isoform.
  • the invention also encompasses other antibodies, preferably monoclonal antibodies or fragments thereof that specifically bind Fc ⁇ RIIB, preferably human Fc ⁇ RIIB, more preferably native human Fc ⁇ RIIB, produced by clones including but not limited to 1D5, 2El, 2H9, 2Dl 1, and 1F2 having ATCC Accession numbers, PTA-5958, PTA-5961, PTA-5962, PTA-5960, PTA-5959, respectively.
  • Hybridomas producing the above-identified clones were deposited with the American Type Culture Collection (10801 University Boulevard., Manassas, VA. 20110-2209) on May 7, 2004, respectively and are incorporated herein by reference.
  • the antibodies of the invention, or fragments thereof agonize at least one activity of Fc ⁇ RIIB.
  • said activity is inhibition of B cell receptor-mediated signaling.
  • the agonistic antibodies of the invention inhibit activation of B cells, B cell proliferation, antibody production, intracellular calcium influx of B cells, cell cycle progression, or activity of one or more downstream signaling molecules in the Fc ⁇ RIIB signal transduction pathway.
  • the agonistic antibodies of the invention enhance phosphorylation of Fc ⁇ RIIB or SHIP recruitment.
  • the agonistic antibodies inhibit MAP kinase activity or Akt recruitment in the B cell receptor-mediated signaling pathway.
  • the agonistic antibodies of the invention agonize Fc ⁇ RIIB -mediated inhibition of Fc ⁇ RI signaling.
  • said antibodies inhibit Fc ⁇ RI-induced mast cell activation, calcium mobilization, degranulation, cytokine production, or serotonin release.
  • the agonistic antibodies of the invention stimulate phosphorylation of Fc ⁇ RIIB, stimulate recruitment of SHIP, stimulate SHIP phosphorylation and its association with She, or inhibit activation of MAP kinase family members (e.g., Erkl, Erk2, JNK, p38, etc.).
  • the agonistic antibodies of the invention enhance tyrosine phosphorylation of p62dok and its association with SHIP and rasGAP. In another embodiment, the agonistic antibodies of the invention inhibit Fc ⁇ R-mediated phagocytosis in monocytes or macrophages.
  • the antibodies of the invention, or fragments thereof antagonize at least one activity of Fc ⁇ RIIB.
  • said activity is activation of B cell receptor-mediated signaling.
  • the antagonistic antibodies of the invention enhance B cell activity, B cell proliferation, antibody production, intracellular calcium influx, or activity of one or more downstream signaling molecules in the Fc ⁇ RIIB signal transduction pathway.
  • the antagonistic antibodies of the invention decrease phosphorylation of Fc ⁇ RIIB or SHIP recruitment.
  • the antagonistic antibodies enhance MAP kinase activity or Akt recruitment in the B cell receptor mediated signaling pathway.
  • the antagonistic antibodies of the invention antagonize Fc ⁇ RIIB -mediated inhibition of Fc ⁇ RI signaling.
  • the antagonistic antibodies of the invention enhance Fc ⁇ RI-induced mast cell activation, calcium mobilization, degranulation, cytokine production, or serotonin release.
  • the antagonistic antibodies of the invention inhibit phosphorylation of Fc ⁇ RIIB, inhibit recruitment of SHIP, inhibit SHIP phosphorylation and its association with She, enhance activation of MAP kinase family members (e.g., Erkl, Erk2, JNK, p38, etc.).
  • the antagonistic antibodies of the invention inhibit tyrosine phosphorylation of p62dok and its association with SHIP and rasGAP. In another embodiment, the antagonistic antibodies of the invention enhance Fc ⁇ R-mediated phagocytosis in monocytes or macrophages. In another embodiment, the antagonistic antibodies of the invention prevent phagocytosis, clearance of opsonized particles by splenic macrophages.
  • Antibodies of the invention include, but are not limited to, monoclonal antibodies, synthetic antibodies, recombinantly produced antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, camelized antibodies, single- chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), intrabodies, and epitope-binding fragments of any of the above.
  • antibodies used in the methods of the present invention include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds to Fc ⁇ RIIB with greater affinity than said immunoglobulin molecule binds Fc ⁇ RIIA or immunospecifically binds Fc ⁇ RIIB and blocks the Fc binding domain of Fc ⁇ RIIB.
  • the antibodies used in the methods of the invention may be from any animal origin including birds and mammals (e.g., human, non-human primate, murine, donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken).
  • the antibodies are human or humanized monoclonal antibodies.
  • "human” antibodies include antibodies
  • NYJD 1603354.2 having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or libraries of synthetic human immunoglobulin coding sequences or from mice that express antibodies from human genes.
  • the antibodies used in the methods of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may immunospecifically bind to different epitopes of Fc ⁇ RIIB or immunospecifically bind to both an epitope of Fc ⁇ RIIB as well a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., International Publication Nos.
  • the antibodies of the invention are multi-specific with specificities for Fc ⁇ RIIB and for a cancer antigen or any other cell surface marker specific for a cell designed to be killed, e.g., in treating or preventing a particular disease or disorder, or for other Fc receptors, e.g., Fc ⁇ RIIIA, Fc ⁇ RIIIB, etc.
  • an antibody used in the methods of the present invention is an antibody or an antigen-binding fragment thereof ⁇ e.g., comprising one or more complementarily determining regions (CDRs), preferably all 6 CDRs) of the antibody produced by clone 2B6 or 3H7 with ATCC accession numbers PTA-4591 and PTA-4592, respectively ⁇ e.g., the heavy chain CDR3).
  • CDRs complementarily determining regions
  • an antibody used in the methods of the present invention binds to the same epitope as the mouse monoclonal antibody produced from clone 2B6 or 3H7 with ATCC accession numbers PTA-4591 and PTA-4592, respectively and/or competes with the mouse monoclonal antibody produced from clone 2B6 or 3H7 with ATCC accession numbers PTA-4591 and PTA-4592, respectively as determined, e.g., in an ELISA assay or other appropriate competitive immunoassay, and also binds Fc ⁇ RIIB with a greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA.
  • the antibodies used in the methods of the invention include derivatives that are modified, Le, by the covalent attachment of any type of molecule to the antibody such that covalent attachment.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to, specific
  • NYJD 16033542 chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids.
  • the derivative may contain one or more non-classical amino acids.
  • human, chimeric or humanized antibodies Completely human antibodies are particularly desirable for therapeutic treatment of human subjects.
  • Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also U.S. Patent Nos. 4,444,887 and 4,716,111; and International Publication Nos.
  • Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes.
  • the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells.
  • the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes.
  • the mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the J H region prevents endogenous antibody production.
  • the modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice.
  • the chimeric mice are then bred to produce homozygous offspring which express human antibodies.
  • the transgenic mice are immunized using conventional methodologies with a selected antigen, e.g., all or a portion of a polypeptide of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • NYJD 1603354.2 Nos. 5,413,923, 5,625,126, 5,633,425, 5,569,825, 5,661,016, 5,545,806, 5,814,318, and 5,939,598, which are incorporated by reference herein in their entirety.
  • companies such as Abgenix, Inc. (Freemont, CA) and Medarex (Princeton, NJ) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different immunoglobulin molecules such as antibodies having a variable region derived from a non-human antibody and a human immunoglobulin constant region.
  • Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, 1985, Science 229:1202; Oi et al, 1986, BioTechniques 4:214; Gillies et al, 1989, J. Immunol. Methods 125:191-202; and U.S. Patent Nos. 6,311,415, 5,807,715, 4,816,567, and 4,816,397, which are incorporated herein by reference in their entirety.
  • Chimeric antibodies comprising one or more CDRs from a non-human species and framework regions from a human immunoglobulin molecule can be produced using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos. 5,225,539, 5,530,101, and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, 1991, Molecular Immunology 28(4/5):489-498; Studnicka et al, 1994, Protein Engineering 7:805; and Roguska et al, 1994, PNAS 91:969), and chain shuffling (U.S. Patent No. 5,565,332).
  • CDR-grafting EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos. 5,225,539, 5,530,101, and 5,585,089)
  • framework residues in the framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. ⁇ See, e.g., U.S. Patent No.
  • a humanized antibody is an antibody, a variant or a fragment thereof which is capable of binding to a predetermined antigen and which comprises a framework region having substantially the amino acid sequence of a human immunoglobulin and a CDR having substantially the amino acid sequence of a non-human immunoglobulin.
  • a humanized antibody comprises substantially all of at least one, and typically two, variable domains in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (Le., donor antibody) and all or substantially all of the framework regions are those of a human
  • a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the antibody will contain both the light chain as well as at least the variable domain of a heavy chain.
  • the antibody also may include the CHl, hinge, CH2, CH3, and CH4 regions of the heavy chain.
  • the humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgG 1 , IgG 2 , IgG 3 and IgG 4 .
  • the constant domain is a complement fixing constant domain where it is desired that the humanized antibody exhibit cytotoxic activity, and the class is typically IgG 1 . Where such cytotoxic activity is not desirable, the constant domain may be of the IgG 2 class.
  • the humanized antibody may comprise sequences from more than one class or isotype, and selecting particular constant domains to optimize desired effector functions is within the ordinary skill in the art.
  • the framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor CDR or the consensus framework may be mutagenized by substitution, insertion or deletion of at least one residue so that the CDR or framework residue at that site does not correspond to either the consensus or the import antibody. Such mutations, however, will not be extensive.
  • humanized antibody residues will correspond to those of the parental framework region (FR) and CDR sequences, more often 90%, and most preferably greater than 95%.
  • Humanized antibodies can be produced using variety of techniques known in the art, including but not limited to, CDR-grafting (European Patent No. EP 239,400; International Publication No. WO 91/09967; and U.S. Patent Nos. 5,225,539, 5,530,101, and 5,585,089), veneering or resurfacing (European Patent Nos.
  • framework residues in the framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by
  • the humanized antibodies of the invention bind the extracellular domain of native human Fc ⁇ RIIB.
  • the humanized anti- Fc ⁇ RIIB antibodies of the invention may have a heavy chain variable region comprising the amino acid sequence of CDRl (SEQ ID NO. 1 or SEQ ID NO. 29) and/or CDR2 (SEQ ID NO.
  • CDR3 SEQ ID NO. 3 or SEQ ID NO. 31
  • a light chain variable region comprising the amino acid sequence of CDRl (SEQ ID NO. 8 or SEQ ID NO. 38) and/or a CDR2 (SEQ ID NO. 9, SEQ ID NO. 10, SEQ ID NO. 11, or SEQ ID NO. 39) and/or CDR3 (SEQ ID NO. 12 or SEQ ID NO. 40).
  • the humanized antibodies of the invention comprise a light chain variable regions comprising an amino acid sequence of SEQ K) NO. 18, SEQ ID NO. 20, SEQ ID NO. 22, or SEQ ID NO. 46, and/or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO. 24 or SEQ ID NO. 37, and/or amino acid sequence variants thereof.
  • the invention encompasses a humanized antibody comprising the CDRs of 2B6 or of 3H7.
  • the invention encompasses a humanized antibody with the heavy chain variable domain having the amino acid sequence of SEQ ID NO: 37 and the light chain variable domain having the amino acid sequence of SEQ ID NO: 46.
  • the invention provides a humanized 2B6 antibody, wherein the VH region consists of the FR segments from the human germline VH segment VH1-18 (Matsuda et al., 1998, J. Exp. Med. 188:2151062) and JH6 (Ravetch et al, 1981, Cell 27(3 Pt. 2): 583-91), and one or more CDR regions of the 2B6 VH, having the amino acid sequence of SED K) NO. 1, SEQ K) NO. 2, or SEQ ID NO. 3.
  • the 2B6 VH has the amino acid sequence of SEQ K) NO. 24.
  • the humanized 2B6 antibody further comprises a VL region, which consists of the FR segments of the human germline VL segment VK- A26 (Lautner-Rieske et al., 1992, Eur. J. Immunol. 22:1023-1029) and JK4 (Hieter et al., 1982, J. Biol. Chem. 257:1516-22), and one or more CDR regions of 2B6VL, having the amino acid sequence of SEQ K ) NO: 8, SEQ K) NO. 9, SEQ K )
  • the 2B6 VL has the amino acid sequence of SEQ ID NO. 18; SEQ ID NO: 20, or SEQ ID NO: 22.
  • the invention provides a humanized 3H7 antibody, wherein the VH region consists of the FR segments from a human germline VH segment and the CDR regions of the 3H7 VH, having the amino acid sequence of SED ID NO. 37.
  • the humanized 3H7 antibody further comprises a VL regions, which consists of the FR segments of a human germline VL segment and the CDR regions of 3H7VL, having the amino acid sequence of SEQ ID NO. 46.
  • the invention provides a humanized antibody that immunospecifically binds to extracellular domain of native human Fc ⁇ RIIB, said antibody comprising (or alternatively, consisting of) CDR sequences of 2B6 or 3H7, in any of the following combinations: a VH CDRl and a VL CDRl; a VH CDRl and a VL CDR2; a VH CDRl and a VL CDR3; a VH CDR2 and a VL CDRl; VH CDR2 and VL CDR2; a VH CDR2 and a VL CDR3; a VH CDR3 and a VH CDRl; a VH CDR3 and a VL CDR2; a VH CDR3 and a VL CDR3; a VHl CDRl, a VH CDR2 and a VL CDRl; a VH CDRl, a VH CDR2 and a VL CDRl; a V
  • At least one CDR from the donor antibody is grafted onto the human antibody.
  • at least two and preferably all three CDRs of each of the heavy and/or light chain variable regions are grafted onto the human antibody.
  • NYJD: 1603354.2 may comprise the Kabat CDRs, the structural loop CDRs or a combination thereof.
  • the invention encompasses a humanized Fc ⁇ RIIB antibody comprising at least one CDR grafted heavy chain and at least one CDR-grafted light chain.
  • the antibodies of the invention can, in turn, be utilized to generate antiidiotype antibodies using techniques well known to those skilled in the art. (See, e.g., Greenspan & Bona, 1989, FASEB J. 7:437-444; and Nissinoff, 1991, /. Immunol. 147:2429- 2438).
  • the invention provides methods employing the use of polynucleotides comprising a nucleotide sequence encoding an antibody of the invention or a fragment thereof.
  • the present invention encompasses single domain antibodies, including camelized single domain antibodies (See e.g., Muyldermans et al., 2001, Trends Biochem. Set 26:230; Nuttall et al, 2000, Cur. Pharm. Biotech. 1:253; Reichmann and Muyldermans, 1999, J. Immunol. Meth. 231:25; International Publication Nos. WO 94/04678 and WO 94/25591; U.S. Patent No. 6,005,079; which are incorporated herein by reference in their entireties).
  • the present invention provides single domain antibodies comprising two VH domains with modifications such that single domain antibodies are formed.
  • the methods of the present invention also encompass the use of antibodies or fragments thereof that have half-lives (e.g., serum half-lives) in a mammal, preferably a human, of greater than 15 days, preferably greater than 20 days, greater than 25 days, greater than 30 days, greater than 35 days, greater than 40 days, greater than 45 days, greater than 2 months, greater than 3 months, greater than 4 months, or greater than 5 months.
  • half-lives e.g., serum half-lives
  • the increased half-lives of the antibodies of the present invention or fragments thereof in a mammal, preferably a human, results in a higher serum titer of said antibodies or antibody fragments in the mammal, and thus, reduces the frequency of the administration of said antibodies or antibody fragments and/or reduces the concentration of said antibodies or antibody fragments to be administered.
  • Antibodies or fragments thereof having increased in vivo half-lives can be generated by techniques known to those of skill in the art. For example, antibodies or fragments thereof with increased in vivo half-lives can be generated by modifying (e.g., substituting, deleting or adding) amino acid residues identified as involved in the interaction between the Fc domain and the FcRn receptor.
  • antibodies of the invention may be engineered by methods described in Ward et al. to increase biological half-lives (See U.S. 6,277,375 Bl).
  • antibodies of the invention may be engineered in the Fc-hinge domain to have increased in vivo or serum half- lives.
  • Antibodies or fragments thereof with increased in vivo half-lives can be generated by attaching to said antibodies or antibody fragments polymer molecules such as high molecular weight polyethyleneglycol (PEG).
  • PEG polymer molecules
  • PEG can be attached to said antibodies or antibody fragments with or without a multifunctional linker either through site-specific conjugation of the PEG to the N- or C- terminus of said antibodies or antibody fragments or via epsilon-amino groups present on lysine residues.
  • Linear or branched polymer derivatization that results in minimal loss of biological activity will be used.
  • the degree of conjugation will be closely monitored by SDS-PAGE and mass spectrometry to ensure proper conjugation of PEG molecules to the antibodies.
  • Unreacted PEG can be separated from antibody-PEG conjugates by, e.g., size exclusion or ion-exchange chromatography.
  • the antibodies of the invention may also be modified by the methods and coupling agents described by Davis et al. (See U.S. 4,179,337) in order to provide compositions that can be injected into the mammalian circulatory system with substantially no immunogenic response.
  • the present invention also encompasses the use of antibodies or antibody fragments comprising the amino acid sequence of any of the antibodies of the invention with mutations (e.g., one or more amino acid substitutions) in the framework or variable regions.
  • mutations in these antibodies maintain or enhance the avidity and/or affinity of the antibodies for the particular antigen(s) to which they immunospecifically bind.
  • Standard techniques known to those skilled in the art e.g., immunoassays
  • the present invention encompasses antibodies comprising modifications preferably, in the Fc region that modify the binding affinity of the antibody to one or more Fc ⁇ R.
  • Methods for modifying antibodies with modified binding to one or more Fc ⁇ R. are known in the art, see, e.g., PCT Publication Nos. WO 99/58572, WO 99/51642, WO 98/23289, WO 89/07142, WO 88/07089, and U.S. Patent Nos. 5,843,597 and 5,642,821, each of which is incorporated herein by reference in their entirety.
  • the invention encompasses any of the mutations disclosed in U.S. Application Nos.
  • the invention encompasses antibodies that have altered affinity for an activating Fc)R, e.g., Fc ⁇ RIIIA. Preferably such modifications also have an altered Fc- mediated effector function. Modifications that affect Fc-mediated effector function are well known in the art (See U.S. 6,194,551, which is incorporated herein by reference in its entirety).
  • NYJD 1603354.2
  • the amino acids that can be modified in accordance with the method of the invention include but are not limited to Proline 329, Proline 331, and Lysine 322.
  • Proline 329, 331 and Lysine 322 are preferably replaced with alanine, however, substitution with any other amino acid is contemplated. See International Publication No.: WO 00/42072 and U.S. 6,194,551 which are incorporated herein by reference in their entirety.
  • the modification of the Fc region comprises one or more mutations in the Fc region.
  • the one or more mutations in the Fc region may result in an antibody with an altered antibody-mediated effector function, an altered binding to other Fc receptors (e.g., Fc activation receptors), an altered ADCC activity, or an altered CIq binding activity, or an altered complement dependent cytotoxicity activity, or any combination thereof.
  • Fc activation receptors e.g., Fc activation receptors
  • ADCC activity e.g., ADCC activity
  • CIq binding activity e.g., CIq binding activity
  • complement dependent cytotoxicity activity e.g., complement dependent cytotoxicity activity
  • Oligosaccharides as used herein refer to carbohydrates containing two or more simple sugars and the two terms may be used interchangeably herein.
  • Carbohydrate moieties of the instant invention will be described with reference to commonly used nomenclature in the art. For a review of carbohydrate chemistry, see, e.g., Hubbard et al, 1981 Ann. Rev. Biochem., 50: 555- 583, which is incorporated herein by reference in its entirety.
  • This nomenclature includes for example, Man which represents mannose; GIcNAc which represents 2-N-acetylglucosamine; Gal which represents galactose; Fuc for fucose and GIc for glucose.
  • Sialic acids are described by the shorthand notation NeuNAc for 5-N-acetylneuraminic acid, and NeuNGc for 5- glycolneuraminic.
  • antibodies contain carbohydrate moeities at conserved positions in the constant region of the heavy chain, and up to 30% of human IgGs have a glycosylated Fab region.
  • IgG has a single N-linked biantennary carbohydrate structure at Asn 297 which resides in the CH2 domain (Jefferis et al, 1998, Immunol. Rev. 163: 59-76; Wright et al, 1997, Trends Biotech 15: 26-32).
  • Human IgG typically has a carbohydrate of the following structure; GlcNAc(Fucose)-GlcNAc-Man-(ManGlcNAc) 2 .
  • the invention encompasses antibodies comprising a variation in the carbohydrate moiety that is attached to Asn 297.
  • the carbohydrate moiety has a galactose and/or galactose-sialic acid at one or both of the terminal GIcNAc and/or a third GlcNac arm (bisecting GIcNAc).
  • the antibodies of the invention are substantially free of one or more selected sugar groups, e.g., one or more sialic acid residues, one or more galactose residues, one or more fucose residues.
  • An antibody that is substantially free of one or more selected sugar groups may be prepared using common methods known to one skilled in the art, including for example recombinantly producing an antibody of the invention in a host cell that is defective in the addition of the selected sugar groups(s) to the carbohydrate moiety of the antibody, such that about 90-100% of the antibody in the composition lacks the selected sugar group(s) attached to the carbohydrate moiety.
  • Alternative methods for preparing such antibodies include for example, culturing cells under conditions which prevent or reduce the addition of one or more selected sugar groups, or post-translational removal of one or more selected sugar groups.
  • the invention encompasses a method of producing a substantially homogenous antibody preparation, wherein about 80-100% of the antibody in the composition lacks a fucose on its carbohydrate moiety, e.g., the carbohydrate attachment on Asn 297.
  • the antibody may be prepared for example by (a) use of an engineered host cell that is deficient in fucose metabolism such that it has a reduced ability to fucosylate proteins expressed therein; (b) culturing cells under conditions which prevent or reduce fusocylation; (c) post- translational removal of fucose, e.g., with a fucosidase enzyme; or (d) purification of the antibody so as to select for the product which is not fucosylated.
  • nucleic acid encoding the desired antibody is expressed in a host cell that has a reduced ability to fucosylate the antibody expressed therein.
  • the host cell is a dihydrofolate reductase deficient Chinese hamster ovary cell (CHO), e.g., a Lee 13 CHO cell (lectin resistant CHO mutant cell line; Ribka & Stanley, 1986, Somatic Cell & Molec. Gen. 12(1): 51-62; Ripka et al., 1986 Arch. Biochem. Biophys. 249(2): 533-45), CHO-Kl, DUX-BI l, CHO-DP12 or CHO-DG44, which has been modified so that the antibody is not substantially fucosylated.
  • CHO dihydrofolate reductase deficient Chinese hamster ovary cell
  • the cell may display altered expression and/or activity for the fucoysltransferase enzyme, or another enzyme or substrate involved in adding fucose to the N-linked oligosaccharide so that the enzyme has a diminished activity and/or reduced expression level in the cell.
  • the fucoysltransferase enzyme or another enzyme or substrate involved in adding fucose to the N-linked oligosaccharide so that the enzyme has a diminished activity and/or reduced expression level in the cell.
  • the altered carbohydrate modifications modulate one or more of the following: solubilization of the antibody, facilitation of subcellular transport and secretion of the antibody, promotion of antibody assembly, conformational integrity, and antibody-mediated effector function.
  • solubilization of the antibody facilitation of subcellular transport and secretion of the antibody
  • promotion of antibody assembly promotion of antibody assembly, conformational integrity, and antibody-mediated effector function.
  • the altered carbohydrate modulate one or more of the following: solubilization of the antibody, facilitation of subcellular transport and secretion of the antibody, promotion of antibody assembly, conformational integrity, and antibody-mediated effector function.
  • the altered carbohydrate modifications modulate one or more of the following: solubilization of the antibody, facilitation of subcellular transport and secretion of the antibody, promotion of antibody assembly, conformational integrity, and antibody-mediated effector function.
  • the altered carbohydrate modulate one or more of the following: solubilization of the antibody, facilitation of subcellular transport and secretion of the antibody, promotion of
  • NYJD 16033542 modifications enhance antibody mediated effector function relative to the antibody lacking the carbohydrate modification.
  • Carbohydrate modifications that lead to altered antibody mediated effector function are well known in the art (for e.g., see Shields R.L. et al., 2001, J. Biol. Chem. 277(30): 26733-40; Davies J. et al, 2001, Biotechnology & Bioengineering, 74(4): 288-294).
  • the altered carbohydrate modifications enhance the binding of antibodies of the invention to Fc ⁇ RIIB receptor.
  • Altering carbohydrate modifications in accordance with the methods of the invention includes, for example, increasing the carbohydrate content of the antibody or decreasing the carbohydrate content of the antibody.
  • the invention encompasses antibodies comprising one or more glycosylation sites, so that one or more carbohydrate moieties are covalently attached to the antibody.
  • the invention encompasses antibodies comprising one or more glycosylation sites and one or more modifications in the Fc region, such as those disclosed supra and those known to one skilled in the art.
  • the one or more modifications in the Fc region enhance the affinity of the antibody for an activating Fc)R, e.g., FcyRIIIA, relative to the antibody comprising the wild type Fc regions.
  • Antibodies of the invention with one or more glycosylation sites and/or one or more modifications in the Fc region have an enhanced antibody mediated effector function, e.g., enhanced ADCC activity.
  • the invention further comprises antibodies comprising one or more modifications of amino acids that are directly or indirectly known to interact with a carbohydrate moiety of the antibody, including but not limited to amino acids at positions 241, 243, 244, 245, 245, 249, 256, 258, 260, 262, 264, 265, 296, 299, and 301.
  • the invention encompasses antibodies that have been modified by introducing one or more glycosylation sites into one or more sites of the antibodies, preferably without altering the functionality of the antibody, e.g., binding activity to Fc)RIIB. Glycosylation sites may be introduced into the variable and/or constant region of the antibodies of the invention. As used herein, "glycosylation sites" include any specific amino acid sequence in an antibody to
  • NYJD 1603354 2 which an oligosaccharide (i.e., carbohydrates containing two or more simple sugars linked together) will specifically and covalently attach.
  • Oligosaccharide side chains are typically linked to the backbone of an antibody via either N-or O-linkages.
  • N-linked glycosylation refers to the attachment of an oligosaccharide moiety to the side chain of an asparagine residue.
  • O- linked glycosylation refers to the attachment of an oligosaccharide moiety to a hydroxyamino acid, e.g., serine, threonine.
  • the antibodies of the invention may comprise one or more glycosylation sites, including N-linked and O-linked glycosylation sites.
  • Any glycosylation site for N-linked or O-linked glycosylation known in the art may be used in accordance with the instant invention.
  • An exemplary N-linked glycosylation site that is useful in accordance with the methods of the present invention is the amino acid sequence: Asn-X-Thr/Ser, wherein X may be any amino acid and Thr/Ser indicates a threonine or a serine.
  • a site or sites may be introduced into an antibody of the invention using methods well known in the art to which this invention pertains. See, for example, "In Vitro Mutagenesis," Recombinant DNA: A Short Course, J. D. Watson, et al. W.H. Freeman and Company, New York, 1983, chapter 8, pp.
  • An exemplary method for introducing a glycosylation site into an antibody of the invention may comprise: modifying or mutating an amino acid sequence of the antibody so that the desired Asn-X-Thr/Ser sequence is obtained.
  • the invention encompasses methods of modifying the carbohydrate content of an antibody of the invention by adding or deleting a glycosylation site.
  • Methods for modifying the carbohydrate content of antibodies are well known in the art and encompassed within the invention, see, e.g., U.S. Patent No. 6,218,149; EP 0 359 096 Bl; U.S. Publication No. US 2002/0028486; WO 03/035835; U.S. Publication No. 2003/0115614; U.S. Patent No. 6,218,149; U.S. Patent No. 6,472,511; all of which are incorporated herein by reference in their entirety.
  • the invention encompasses methods of modifying the carbohydrate content of an antibody of the invention by deleting one or more endogenous carbohydrate moieties of the antibody.
  • the invention further encompasses methods of modifying an effector function of an antibody of the invention, wherein the method comprises modifying the carbohydrate content of the antibody using the methods disclosed herein or known in the art.
  • Standard techniques known to those skilled in the art can be used to introduce mutations in the nucleotide sequence encoding an antibody, or fragment thereof, including, e.g., site-directed mutagenesis and PCR-mediated mutagenesis, which results in amino acid substitutions.
  • the derivatives include less than 15 amino acid substitutions, less than
  • NYJD 16033542 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the original antibody or fragment thereof.
  • the derivatives have conservative amino acid substitutions made at one or more predicted non-essential amino acid residues.
  • the present invention also encompasses antibodies or fragments thereof comprising an amino acid sequence of a variable heavy chain and/or variable light chain that is at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% identical to the amino acid sequence of the variable heavy chain and/or light chain of the mouse monoclonal antibody produced by clone 2B6 or 3H7 having ATCC accession numbers PTA-4591 and PTA-4592, respectively.
  • the present invention further encompasses antibodies or fragments thereof that specifically bind Fc ⁇ RIIB with greater affinity than said antibody or fragment thereof binds Fc ⁇ RIIA and antibodies or a fragments thereof that specifically binds Fc ⁇ RIIB and block the Fc binding domain of Fc ⁇ RIIB, said antibodies or antibody fragments comprising an amino acid sequence of one or more CDRs that is at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% identical to the amino acid sequence of one or more CDRs of the mouse monoclonal antibody produced by clone 2B6 or 3H7 having ATCC accession numbers PTA-4591 and PTA- 4592, respectively.
  • the determination of percent identity of two amino acid sequences can be determined by any method known to one skilled in the art, including BLAST protein searches.
  • the present invention also encompasses the use of antibodies or antibody fragments that specifically bind Fc ⁇ RIIB with greater affinity than said antibodies or fragments thereof binds Fc ⁇ RIIA and antibodies or antibody fragments thereof that specifically binds Fc ⁇ RIIB and block the Fc binding domain of Fc ⁇ RIIB, wherein said antibodies or antibody fragments are encoded by a nucleotide sequence that hybridizes to the nucleotide sequence of the mouse monoclonal antibody produced by clone 2B6 or 3H7 having ATCC accession numbers PTA-4591 and PTA-4592, respectively, under stringent conditions.
  • the invention provides antibodies or fragments thereof that specifically bind Fc ⁇ RIIB with greater affinity than said antibodies or fragments thereof bind Fc ⁇ RIIA and antibodies or a fragments thereof that specifically binds Fc ⁇ RIIB and block the Fc binding domain of Fc ⁇ RIIB, said antibodies or antibody fragments comprising a variable light and/or variable heavy chain encoded by a nucleotide sequence that hybridizes under stringent conditions to the nucleotide sequence of the variable light and/or variable heavy chain of the
  • the invention provides antibodies or fragments thereof that specifically bind Fc ⁇ RIIB with greater affinity than said antibodies or fragments thereof bind Fc ⁇ RIIA and antibodies or a fragments thereof that specifically binds Fc ⁇ RIIB and block the Fc binding domain of Fc ⁇ RIIB, said antibodies or antibody fragments comprising one or more CDRs encoded by a nucleotide sequence that hybridizes under stringent conditions to the nucleotide sequence of one or more CDRs of the mouse monoclonal antibody produced by clone 2B6 or 3H7 with ATCC accession numbers PTA-4591 and PTA-4592, respectively.
  • Stringent hybridization conditions include, but are not limited to, hybridization to filter-bound DNA in 6X sodium chloride/sodium citrate (SSC) at about 45 0 C followed by one or more washes in 0.2X SSC/0.1% SDS at about 50-65°C, highly stringent conditions such as hybridization to filter- bound DNA in 6X SSC at about 45°C followed by one or more washes in 0.1X SSC/0.2% SDS at about 60 0 C, or any other stringent hybridization conditions known to those skilled in the art ⁇ see, for example, Ausubel, F.M. et al, eds. 1989 Current Protocols in Molecular Biology, vol. 1, Green Publishing Associates, Inc. and John Wiley and Sons, Inc., NY at pages 6.3.1 to 6.3.6 and 2.10.3), incorporated herein by reference.
  • SSC 6X sodium chloride/sodium citrate
  • the present invention encompasses antibodies recombinantly fused or chemically conjugated (including both covalently and non-covalently conjugations) to heterologous polypeptides ⁇ i.e., an unrelated polypeptide; or portion thereof, preferably at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids of the polypeptide) to generate fusion proteins.
  • the fusion does not necessarily need to be direct, but may occur through linker sequences.
  • Antibodies may be used for example to target heterologous polypeptides to particular cell types, either in vitro or in vivo, by fusing or conjugating the antibodies to antibodies specific for particular cell surface receptors.
  • Antibodies fused or conjugated to heterologous polypeptides may also be used in in vitro immunoassays and purification methods using methods known in the art. See e.g., PCT publication Number WO 93/2 1232; EP 439,095; Naramura et al, Immunol. Lett, 39:91-99, 1994; U.S. Patent 5,474,981; Gillies et al, PNAS, 89:1428-1432, 1992; and Fell et al., J. Immunol., 146:2446- 2452, 1991, which are incorporated herein by reference in their entireties. [00142] Further, an antibody may be conjugated to a therapeutic agent or drug moiety that modifies a given biological response. Therapeutic agents or drug moieties are not to be
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin (i.e., PE-40), or diphtheria toxin, ricin, gelonin, and pokeweed antiviral protein, a protein such as tumor necrosis factor, interferons including, but not limited to, ⁇ -interferon (IFN- ⁇ ), ⁇ -interferon (IFN- ⁇ ), nerve growth factor (NGF), platelet derived growth factor (PDGF), tissue plasminogen activator (TPA), an apoptotic agent (e.g., TNF- ⁇ , TNF- ⁇ , AIM I as disclosed in PCT Publication No.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin (i.e., PE-40), or diphtheria toxin, ricin, gelonin, and pokeweed
  • a thrombotic agent or an anti-angiogenic agent e.g., angiostatin or endostatin
  • a biological response modifier such as, for example, a lymphokine (e.g., interleukin-1 ("IL-I”), interleukin-2 ("IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), and granulocyte colony stimulating factor (“G-CSF')), macrophage colony stimulating factor, (“M- CSF”), or a growth factor (e.g., growth hormone (“GH”); proteases, or ribonucleases.
  • IL-I interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • G-CSF' granulocyte colony stimulating factor
  • M- CSF macrophage colony stimulating factor
  • M- CSF growth factor
  • GH growth hormone
  • Antibodies can be fused to marker sequences, such as a peptide to facilitate purification
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • a pQE vector QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311
  • hexa- histidine provides for convenient purification of the fusion protein.
  • peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell, 37:767 1984) and the "flag” tag (Knappik et al., Biotechniques, 17(4):754-761, 1994).
  • HA hemagglutinin
  • the present invention further includes compositions comprising heterologous polypeptides fused or conjugated to antibody fragments.
  • the heterologous polypeptides may be fused or conjugated to a Fab fragment, Fd fragment, Fv fragment, F(ab) 2 fragment, or portion thereof.
  • Methods for fusing or conjugating polypeptides to antibody portions are known in the art. See, e.g., U.S. Patent Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, and 5,112,946; EP 307,434; EP 367,166; International Publication Nos.
  • Additional fusion proteins may be generated through the techniques of gene- shuffling, motif-shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as
  • DNA shuffling may be employed to alter the activities of antibodies of the invention or fragments thereof ⁇ e.g., antibodies or fragments thereof with higher affinities and lower dissociation rates). See, generally, U.S. Patent Nos. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and Patten et al, 1997, Curr. Opinion Biotechnol. 8:724-33; Harayama, 1998, Trends Biotechnol. 16:76; Hansson, et al, 1999, J. MoI. Biol.
  • Antibodies or fragments thereof, or the encoded antibodies or fragments thereof may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination.
  • One or more portions of a polynucleotide encoding an antibody or antibody fragment, which portions specifically bind to Fc ⁇ RIIB may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous molecules.
  • the present invention also encompasses antibodies conjugated to a diagnostic or therapeutic agent or any other molecule for which serum half-life is desired to be increased.
  • the antibodies can be used diagnostically to, for example, monitor the development or progression of a disease, disorder or infection as part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals, and nonradioactive paramagnetic metal ions.
  • the detectable substance may be coupled or conjugated either directly to the antibody or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention.
  • Such diagnosis and detection can be accomplished by coupling the antibody to detectable substances including, but not limited to, various enzymes, enzymes including, but not limited to, horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; prosthetic group complexes such as, but not limited to, streptavidin/biotin and avidin/biotin; fluorescent materials such as, but not limited to, umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent material such as, but not limited to, luminol; bioluminescent materials such as, but not limited to, luciferase, luciferin, and aequorin; radioactive material such as, but not limited to, bismuth ( 213 Bi), carbon ( 14 C), chromium ( 51 Cr),
  • NYJD 16Q3354.2 67 Ga), germanium ( 68 Ge), holmium ( 166 Ho), indium ( 1 15 In, 1 13 In, 1 12 In, 1 11 In), iodine ( 131 I, 125 I, 123 1, 121 I), lanthanium ( 140 La), lutetium ( 177 Lu), manganese ( 54 Mn), molybdenum ( 99 Mo), palladium ( 103 Pd), phosphorous ( 32 P), praseodymium ( 142 Pr), promethium ( 149 Pm), rhenium ( 186 Re, 188 Re), rhodium ( 105 Rh), ruthemium ( 97 Ru), samarium ( 153 Sm), scandium ( 47 Sc), selenium ( 75 Se), strontium ( 85 Sr), sulfur ( 35 S), technetium ( 99 Tc), thallium ( 201 Ti), tin ( 113 Sn, 117 Sn), tritium ( 3 H), xenon ( 133
  • An antibody may be conjugated to a therapeutic moiety such as a cytotoxin (e.g., a cytostatic or cytocidal agent), a therapeutic agent or a radioactive element (e.g., alpha-emitters, gamma-emitters, etc.).
  • cytotoxin e.g., a cytostatic or cytocidal agent
  • therapeutic agent e.g., a therapeutic agent
  • a radioactive element e.g., alpha-emitters, gamma-emitters, etc.
  • Cytotoxins or cytotoxic agents include any agent that is detrimental to cells.
  • Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cisdichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g.,
  • an antibody can be conjugated to therapeutic moieties such as a radioactive materials or macrocyclic chelators useful for conjugating radiometal ions (see above for examples of radioactive materials).
  • the macrocyclic chelator is l,4,7,10-tetraazacyclododecane-N,N',N",N"-tetraacetic acid (DOTA) which can be attached to the antibody via a linker molecule.
  • linker molecules are commonly known in the art and described in Denardo et al, 1998, Clin Cancer Res. 4:2483-90; Peterson et al, 1999, Bioconjug. Chem. 10:553; and Zimmerman et al, 1999, Nucl. Med. Biol. 26:943-50 each incorporated by reference in their entireties.
  • An antibody or fragment thereof, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980, which is incorporated herein by reference in its entirety.
  • Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al, Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al, in: Monoclonal Antibodies and T-CeIl Hvbridomas, pp. 563-681 (Elsevier, N. Y., 1981) (both of which are incorporated by reference in their entireties).
  • the term “monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • mice can be immunized with an antigen of interest or a cell expressing such an antigen.
  • NYJD 1603354 2 response is detected, e.g., antibodies specific for the antigen are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated. The splenocytes are then fused by well known techniques to any suitable myeloma cells. Hybridomas are selected and cloned by limiting dilution. The hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding the antigen. Ascites fluid, which generally contains high levels of antibodies, can be generated by inoculating mice intraperitoneally with positive hybridoma clones.
  • the invention provides a method for producing monoclonal antibodies that specifically bind Fc ⁇ RIIB with greater affinity than said monoclonal antibodies bind Fc ⁇ RIIA comprising: immunizing one or more Fc ⁇ RIIA transgenic mice (See U.S. 5,877,396 and U.S. 5,824,487) with the purified extracellular domain of human Fc ⁇ RIIB, amino acids 1-180; producing hybridoma cell lines from spleen cells of said mice, screening said hybridoma cells lines for one or more hybridoma cell lines that produce antibodies that specifically bind Fc ⁇ RIIB with greater affinity than said antibodies bind Fc ⁇ RIIA.
  • the invention provides a method for producing Fc ⁇ RIIB monoclonal antibodies that specifically bind Fc ⁇ RIIB, particularly human Fc ⁇ RIIB, with a greater affinity than said monoclonal antibodies bind Fc ⁇ RIIA, said method further comprising: immunizing one or more Fc ⁇ RIIA transgenic mice with purified Fc ⁇ RIIB or an immunogenic fragment thereof, booster immunizing said mice sufficient number of times to elicit an immune response, producing hybridoma cells lines from spleen cells of said one or more mice, screening said hybridoma cell lines for one or more hybridoma cell lines that produce antibodies that specifically bind Fc ⁇ RIIB with a greater affinity than said antibodies bind Fc ⁇ RIIA.
  • said mice are immunized with purified Fc ⁇ RIIB which has been mixed with any adjuvant known in the art to enhance immune response.
  • adjuvants that can be used in the methods of the invention include, but are not limited to, protein adjuvants; bacterial adjuvants, e.g., whole bacteria (BCG, Corynebacterium parvum, Salmonella minnesota) and bacterial components including cell wall skeleton, trehalose dimycolate, monophosphoryl lipid A, methanol extractable residue (MER) of tubercle bacillus, complete or incomplete Freund's adjuvant; viral adjuvants; chemical adjuvants, e.g., aluminum hydroxide, iodoacetate and cholesteryl hemisuccinateor; naked DNA adjuvants.
  • BCG whole bacteria
  • Corynebacterium parvum Salmonella minnesota
  • bacterial components including cell wall skeleton, trehalose dimycolate, monophosphoryl
  • adjuvants that can be used in the methods of the invention include, Cholera toxin, paropox proteins, MF-59 (Chiron Corporation; See also Bieg et al, 1999, Autoimmunity, 31(l):15-24, which is incorporated herein by reference), MPL® (Corixa Corporation; See also Lodmell D.I. et al, 2000 Vaccine, 18: 1059- 1066; Ulrich et al, 2000, Methods in Molecular Medicine, 273-282; Johnson et al, 1999,
  • Antibody fragments which recognize specific epitopes may be generated by known techniques.
  • Fab and F(ab') 2 fragments may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab') 2 fragments).
  • F(ab') 2 fragments contain the complete light chain, and the variable region, the CHl region and the hinge region of the heavy chain.
  • antibodies can also be generated using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen binding domains, such as Fab and Fv or disulfide-bond stabilized Fv, expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage, including fd and M13.
  • the antigen binding domains are expressed as a recombinantly fused protein to either the phage gene HI or gene VIII protein.
  • phage display methods that can be used to make the immunoglobulins, or fragments thereof, of the present invention include those disclosed in Brinkman et al, J. Immunol. Methods, 182:41- 50, 1995; Ames et al, J. Immunol Methods, 184:177-186, 1995; Kettleborough et al, Eur. J. Immunol, 24:952-958, 1994; Persic et al, Gene, 187:9-18, 1997; Burton et al, Advances in Immunology, 57:191-280, 1994; PCT application No.
  • NYJD 1603354.2 mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below.
  • techniques to recombinantly produce Fab, Fab' and F(ab') 2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et al, BioTechniques, 12(6): 864-869, 1992; and Sawai et al., AJRI, 34:26- 34, 1995; and Better et al, Science, 240: 1041-1043, 1988 (each of which is incorporated by reference in its entirety).
  • Phage display technology can be used to increase the affinity of an antibody of the invention for Fc ⁇ RIIB. This technique would be useful in obtaining high affinity antibodies that could be used in the combinatorial methods of the invention.
  • the technology referred to as affinity maturation, employs mutagenesis or CDR walking and re-selection using Fc ⁇ RIIB or an antigenic fragment thereof to identify antibodies that bind with higher affinity to the antigen when compared with the initial or parental antibody (See, e.g.,Glaser et al, 1992, J. Immunology 149:3903). Mutagenizing entire codons rather than single nucleotides results in a semi- randomized repertoire of amino acid mutations.
  • Libraries can be constructed consisting of a pool of variant clones each of which differs by a single amino acid alteration in a single CDR and which contain variants representing each possible amino acid substitution for each CDR residue.
  • Mutants with increased binding affinity for the antigen can be screened by contacting the immobilized mutants with labeled antigen. Any screening method known in the art can be used to identify mutant antibodies with increased- avidity to the antigen (e.g., ELISA) (See Wu et al, 1998, Proc Natl Acad Sci. USA 95:6037; Yelton et al, 1995, J. Immunology 155:1994).
  • CDR walking which randomizes the light chain is also possible (See Schier et al, 1996, J. MoI Bio. 263:551).
  • Antibodies of the invention may be further characterized by epitope mapping, so that antibodies may be selected that have the greatest specificity for Fc ⁇ RIIB compared to Fc ⁇ RIIA.
  • Epitope mapping methods of antibodies are well known in the art and encompassed within the methods of the invention.
  • fusion proteins comprising one or more regions of Fc ⁇ RIIB may be used in mapping the epitope of an antibody of the invention.
  • the fusion protein contains the amino acid sequence of a region of an Fc ⁇ RIIB fused to the Fc portion of human IgG2.
  • Each fusion protein may further comprise amino acid substitutions and/or replacements of certain regions of the receptor with the
  • NYJD 1603354.2 corresponding region from a homolog receptor, e.g., Fc)RIIA, as shown in Table 2 below.
  • pMGX125 and pMGX132 contain the IgG binding site of the FcyRIIB receptor, the former with the C-terminus of Fc ⁇ RIIB and the latter with the C-terminus of Fc ⁇ RIIA and can be used to differentiate C-terminus binding.
  • the others have Fc ⁇ RIIA substitutions in the IgG binding site and either the Fc ⁇ llA or Fc ⁇ llB N-terminus. These molecules can help determine the part of the receptor molecule where the antibodies bind.
  • the fusion proteins may be used in any biochemical assay for determination of binding to an anti-Fc ⁇ RIIB antibody of the invention, e.g., an ELISA.
  • further confirmation of the epitope specificity may be done by using peptides with specific residues replaced with those from the Fc ⁇ RIIA sequence.
  • the antibodies of the invention may be characterized for specific binding to
  • Fc ⁇ RIIB using any immunological or biochemical based method known in the art for characterizing including quantitating, the interaction of the antibody to Fc ⁇ RIIB.
  • Specific binding of an antibody of the invention to Fc ⁇ RIIB may be determined for example using immunological or biochemical based methods including, but not limited to, an ELISA assay, surface plasmon resonance assays, immunoprecipitation assay, affinity chromatography, and equilibrium dialysis. Immunoassays which can be used to analyze immunospecific binding and
  • cross-reactivity of the antibodies of the invention include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, to name but a few.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradi
  • Antibodies of the invention may also be assayed using any surface plasmon resonance based assays known in the art for characterizing the kinetic parameters of the interaction of the antibody with Fc ⁇ RIIB.
  • Any SPR instrument commercially available including, but not limited to, BIAcore Instruments, available from Biacore AB (Uppsala, Sweden); IAsys instruments available from Affinity Sensors (Franklin, MA.); IBIS system available from Windsor Scientific Limited (Berks, UK), SPR-CELLLA systems available from Nippon Laser and Electronics Lab (Hokkaido, Japan), and SPR Detector Spreeta available from Texas Instruments (Dallas, TX) can be used in the instant invention.
  • SPR based assays involve immobilizing a member of a binding pair on a surface, and monitoring its interaction with the other member of the binding pair in solution in real time.
  • SPR is based on measuring the change in refractive index of the solvent near the surface that occurs upon complex formation or dissociation.
  • the surface onto which the immobilization occur is the sensor chip, which is at the heart of the SPR technology; it consists of a glass surface coated with a thin layer of gold and forms the basis for a range of specialized surfaces designed to optimize the binding of a molecule to the surface.
  • a variety of sensor chips are commercially available especially from the companies listed supra, all of which may be used in the methods of the invention. Examples of sensor chips include those available from BIAcore
  • a molecule of the invention may be immobilized onto the surface of a sensor chip using any of the immobilization methods and chemistries known in the art, including but not limited to, direct covalent coupling via amine groups, direct covalent coupling via sulfhydryl groups, biotin attachment to avidin coated surface, aldehyde coupling to carbohydrate groups, and attachment through the histidine tag with NTA chips.
  • the invention encompasses characterization of the antibodies produced by the methods of the invention using certain characterization assays for identifying the function of the antibodies of the invention, particularly the activity to modulate Fc ⁇ RIIB signaling.
  • characterization assays of the invention can measure phosphorylation of tyrosine residues in the ITIM motif of Fc ⁇ RIIB, or measure the inhibition of B cell receptor- generated calcium mobilization.
  • the characterization assays of the invention can be cell-based or cell-free assays.
  • Fc ⁇ RIIB is rapidly phosphorylated on tyrosine in its ITIM motif, and then recruits Src Homology-2 containing inositol-5-phosphatase (SHIP), an SH2 domain- containing inosital polyphosphate 5-phosphatase, which is in turn phosphorylated and associates with She and p62 dok
  • SHIP inositol-5-phosphatase
  • p62 dok is the prototype of a family of adaptor molecules, which includes signaling domains such as an aminoterminal pleckstrin homology domain (PH domain), a PTB domain, and a carboxy terminal region containing PXXP motifs and numerous phosphorylation sites (Carpino et al., 1997 Cell, 88: 197; Yamanshi et al, 1997, Cell, 88:205).
  • the invention encompasses characterizing the anti-Fc ⁇ RIIB antibodies of the invention in modulating one or more IgE mediated responses.
  • cells lines co- expressing the high affinity receptor for IgE and the low affinity receptor for Fc ⁇ RIIB will be used in characterizing the anti-Fc ⁇ RIIB antibodies of the invention in modulating IgE mediated responses.
  • cells from a rat basophilic leukemia cell line (RBL-H23; Barsumian E.L. et al. , 1981, EMr. J. Immunol.11:317, which is incorporated herein by reference in its entirety) transfected with full length human Fc ⁇ RIIB will be used in the methods of the invention.
  • RBL-2H3 is a well characterized rat cell line that has been used extensively to study
  • NYJD 1603354.2 the signaling mechanisms following IgE-mediated cell activation.
  • Fc ⁇ RIIB inhibits Fc ⁇ RI-induced calcium mobilization, degranulation, and cytokine production (Malbec et al, 1998, J. Immunol. 160:1647; Daeron et al, 1995 J. Clin. Invest. 95:577; Ott et al, 2002 J. of Immunol. 168:4430-4439).
  • the invention encompasses characterizing the anti-
  • Fc ⁇ RIIB antibodies of the invention for inhibition of Fc ⁇ RI induced mast cell activation.
  • cells from a rat basophilic leukemia cell line RBL-H23; Barsumian E.L. et al. 1981 EMr. J. Immunol. ⁇ :3 ⁇ l
  • Fc ⁇ RIIB antibodies of the invention for inhibition of Fc ⁇ RI induced mast cell activation.
  • cells from a rat basophilic leukemia cell line RBL-H23; Barsumian E.L. et al. 1981 EMr. J. Immunol. ⁇ :3 ⁇ l
  • Fc ⁇ RIIB rat basophilic leukemia cell line
  • indirect modulation of down stream signaling molecules can be assayed upon addition of antibodies of the invention to the sensitized and stimulated cells.
  • tyrosine phosphorylation of Fc ⁇ RIIB and recruitment and phosphorylation of SHIP activation of MAP kinase family members, including but not limited to Erkl, Erk2, JNK, or p38; and tyrosine phosphorylation of p62 dok and its association with SHIP and RasGAP can be assayed.
  • One exemplary assay for determining the inhibition of Fc ⁇ RI induced mast cell activation by the antibodies of the invention can comprise of the following: transfecting RBL- H23 cells with human Fc ⁇ RIIB; sensitizing the RBL-H23 cells with IgE; stimulating RBL-H23 cells with either F(ab') 2 of rabbit anti-mouse IgG (to aggregate Fc ⁇ RI alone and elicit Fc ⁇ RI- mediated signaling, as a control), or stimulating RBL-H23 cells with whole rabbit anti-mouse IgG to (to coaggregate Fc ⁇ RIIB and Fc ⁇ RI, resulting in inhibition of Fc ⁇ RI-mediated signaling).
  • Cells that have been stimulated with whole rabbit anti-mouse IgG antibodies can be further pre- incubated with the antibodies of the invention. Measuring Fc ⁇ RI-dependent activity of cells that have been pre-incubated with the antibodies of the invention and cells that have not been pre- incubated with the antibodies of the invention, and comparing levels of Fc ⁇ RI-dependent activity in these cells, would indicate a modulation of Fc ⁇ RI-dependent activity by the antibodies of the invention.
  • the exemplary assay described above can be for example, used to identify antibodies that block ligand (IgG) binding to Fc ⁇ RIIB receptor and antagonize Fc ⁇ RIIB - mediated inhibition of Fc ⁇ RI signaling by preventing coaggregating of Fc ⁇ RIIB and Fc ⁇ RI.
  • This assay likewise identifies antibodies that enhance coaggregation of Fc ⁇ RIIB and Fc ⁇ RI and agonize Fc ⁇ RIIB -mediated inhibition of Fc ⁇ RI signaling by promoting coaggregating of Fc ⁇ RIIB and Fc ⁇ RI.
  • Fc ⁇ RI-dependent activity is at least one or more of the following: modulation of downstream signaling molecules (e.g., modulation of phosphorylation state of Fc ⁇ RIIB, modulation of SHIP recruitment, modulation of MAP Kinase activity, modulation of phosphorylation state of SHIP, modulation of SHIP and She association SHIP and She, modulation of the phosphorylation state of p62 dok , modulation of p62 dok and SHIP association, modulation of p62 dok and RasGAP association, modulation of calcium mobilization, modulation of degranulation, and modulation of cytokine production.
  • modulation of downstream signaling molecules e.g., modulation of phosphorylation state of Fc ⁇ RIIB, modulation of SHIP recruitment, modulation of MAP Kinase activity, modulation of phosphorylation state of SHIP, modulation of SHIP and She association SHIP and She, modulation of the phosphorylation state of p62 dok , modulation of p62
  • Fc ⁇ RI-dependent activity is serotonin release and/or extracellular Ca ++ influx and/or IgE dependent mast cell activation. It is known to one skilled in the art that coaggregation of Fc ⁇ RIIB and Fc ⁇ RI stimulates Fc ⁇ RIIB tyrosine phosphorylation, stimulates recruitment of SHIP, stimulates SHIP tyrosine phosphorylation and association with She, and inhibits activation of MAP kinase family members including, but not limited to, Erkl, Erk2, JNK, p38. It is also known to those skilled in the art that coaggregation of Fc ⁇ RIIB and Fc ⁇ RI stimulates enhanced tyrosine phosphorylation of p62 dok and its association with SHIP and RasGAP.
  • the anti-Fc ⁇ RIIB antibodies of the invention are characterized for their ability to modulate an IgE mediated response by monitoring and/or measuring degranulation of mast cells or basophils, preferably in a cell-based assay.
  • mast cells or basophils for use in such assays have been engineered to contain human Fc ⁇ RIIB using standard recombinant methods known to one skilled in the art.
  • the anti-Fc ⁇ RIIB antibodies of the invention are characterized for their ability to modulate an IgE mediated response in a cell-based ⁇ -hexosaminidase (enzyme contained in the granules) release assay, ⁇ -hexosaminidase release from mast cells and basophils is a primary event in acute allergic and inflammatory condition (Aketani et ai, 2001 Immunol. Lett. 75: 185-9; Aketani et al., 2000 Anal. Chem. 72: 2653-8). Release of other inflammatory mediators including but not limited to serotonin and histamine may be assayed to measure an IgE mediated response in accordance with the methods of the invention.
  • ⁇ -hexosaminidase release assay comprising the following: transfecting RBL-H23 cells with human Fc ⁇ RIIB; sensitizing the cells
  • NYJD- 16033542 with mouse IgE alone or with mouse IgE and an anti-Fc ⁇ RIIB antibody of the invention; stimulating the cells with various concentrations of goat anti-mouse F(ab) 2 , preferably in a range from 0.03 ⁇ g/mL to 30 ⁇ g/mL for about 1 hour; collecting the supernatant; lysing the cells; and measuring the ⁇ -hexosaminidase activity released in the supernatant by a colorometric assay, e.g., using p-nitrophenyl N-acetyl- ⁇ -D-glucosaminide. The released ⁇ -hexosaminidase activity is expressed as a percentage of the released activity to the total activity.
  • the released ⁇ - hexosaminidase activity will be measured and compared in cells treated with antigen alone; IgE alone; IgE and an anti-Fc ⁇ RIIB antibody of the invention.
  • antigen alone IgE alone
  • IgE and an anti-Fc ⁇ RIIB antibody of the invention an antigen of the invention.
  • aggregation and cross linking of Fc ⁇ RI occurs since the polyclonal antibody recognizes the light chain of the murine IgE bound to the Fc ⁇ RI; which in turn leads to mast cell activation and degranulation.
  • the anti-Fc ⁇ RIIB antibodies bound to the Fc ⁇ RIIB receptor and cross linked to Fc ⁇ RI do not affect the activation of the inhibitory pathway, i.e., there is no alteration in the level of degranulation in the presence of an anti-Fc ⁇ RIIB antibody.
  • the anti-Fc ⁇ RIIB antibodies mediate a stronger activation of the inhibitory receptor, Fc ⁇ RIIB, when bound by the anti-Fc ⁇ RIIB antibody, allowing effective cross linking to Fc ⁇ RI and activation of the inhibitory pathway of homo- aggregated Fc ⁇ RIIB.
  • the invention also encompasses characterizing the effect of the anti-Fc ⁇ RIIB antibodies of the invention on IgE mediated cell response using calcium mobilization assays using methodologies known to one skilled in the art.
  • An exemplary calcium mobilization assay may comprise the following: priming basophils or mast cells with IgE; incubating the cells with a calcium indicator, e.g., Fura 2; stimulating cells as described supra; and monitoring and/or quantitating intracellular calcium concentration for example by using flow cytometry.
  • the invention encompasses monitoring and/or quantitating intracellular calcium concentration by any method known to one skilled in the art see, e.g., Immunology Letters, 2001, 75:185-9; British J. ofPharm, 2002, 136:837-45; J. of Immunology, 168:4430-9 and /, of Cell Biol, 153(2):339-49; all of which are incorporated herein by reference.
  • anti-Fc ⁇ RIIB antibodies of the invention inhibit IgE mediated cell activation.
  • the anti-Fc ⁇ RIIB antibodies of the invention block the inhibitory pathways regulated by Fc ⁇ RIIB or block the ligand binding site on Fc ⁇ RIIB and thus enhance immune response.
  • LAD 1 and LAD2 novel stem cell factor dependent human mast cell lines
  • Both cell lines have been described to express Fc ⁇ RI and several human mast cell markers.
  • the invention encompasses using LAD 1 and 2 cells in the methods of the invention for assessing the effect of the antibodies of the invention on IgE mediated responses.
  • cell-based ⁇ - hexosaminidase release assays such as those described supra may be used in LAD cells to determine any modulation of the IgE-mediated response by the anti-Fc ⁇ RIIB antibodies of the invention.
  • human mast cells e.g., LAD 1
  • LAD 1 are primed with chimaeric human IgE anti-nitrophenol (NP) and challenged with BSA-NP, the polyvalent antigen, and cell degranulation is monitored by measuring the ⁇ -hexosaminidase released in the supernatant (Kirshenbaum et al, 2003, Leukemia research, 27:677-682, which is incorporated herein by reference in its entirety).
  • Fc ⁇ RIIB as determined using standard methods known in the art, e.g., FACS staining, it may be difficult to monitor and/or detect differences in the activation of the inhibitory pathway mediated by the anti-Fc ⁇ RIIB antibodies of the invention.
  • the invention thus encompasses alternative methods, whereby the Fc ⁇ RIIB expression may be upregulated using cytokines and particular growth conditions.
  • Fc ⁇ RIIB has been described to be highly up-regulated in human monocyte cell lines, e.g., THPl and U937, (Tridandapani et al, 2002, J. Biol. Chem., 277(7): 5082-5089) and in primary human monocytes (Pricop et al., 2001, J.
  • the invention also encompasses characterizing the anti-Fc ⁇ RIIB antibodies of the invention for inhibition of B-cell receptor (BCR)-mediated signaling.
  • BCR-mediated signaling can include at least one or more down stream biological responses, such as activation and proliferation of B cells, antibody production, etc.
  • Coaggregation of Fc ⁇ RIIB and BCR leads to inhibition of cell cycle progression and cellular survival. Further, coaggregation of Fc ⁇ RIIB and BCR leads to inhibition of BCR-mediated signaling.
  • BCR-mediated signaling comprises at least one or more of the following: modulation of down stream signaling molecules ⁇ e.g., phosphorylation state of Fc ⁇ RIIB, SHIP recruitment, localization of Btk and/or PLC ⁇ , MAP kinase activity, recruitment of Akt (anti-apoptotic signal), calcium mobilization, cell cycle progression, and cell proliferation.
  • modulation of down stream signaling molecules ⁇ e.g., phosphorylation state of Fc ⁇ RIIB, SHIP recruitment, localization of Btk and/or PLC ⁇
  • MAP kinase activity e.g., MAP kinase activity
  • Akt anti-apoptotic signal
  • One exemplary assay for determining Fc ⁇ RIIB -mediated inhibition of BCR signaling by the antibodies of the invention can comprise the following: isolating splenic B cells from SHIP deficient mice, activating said cells with lipopolysachharide, and stimulating said cells with either F(ab') 2 anti-IgM to aggregate BCR or with anti-IgM to coaagregate BCR with Fc ⁇ RIIB.
  • Cells that have been stimulated with intact anti-IgM to coaggregate BCR with Fc ⁇ RIIB can be further pre-incubated with the antibodies of the invention.
  • Fc ⁇ RIIB -dependent activity of cells can be measured by standard techniques known in the art.
  • Measuring Fc ⁇ RIIB -dependent activity can include, for example, measuring intracellular calcium mobilization by flow cytometry, measuring phosphorylation of Akt and/or Erk, measuring BCR-mediated accumulation of PI(3,4,5)P 3 , or measuring Fc ⁇ RIIB -mediated proliferation B cells.
  • the assays can be used, for example, to identify antibodies that modulate
  • Fc ⁇ RIIB-mediated inhibition of BCR signaling by blocking the ligand (IgG) binding site to Fc ⁇ RIIB receptor and antagonizing Fc ⁇ RIIB-mediated inhibition of BCR signaling by
  • NYJD 1603354.2 preventing coaggregation of Fc ⁇ RIIB and BCR.
  • the assays can also be used to identify antibodies that enhance coaggregation of Fc ⁇ RIIB and BCR and agonize Fc ⁇ RIIB -mediated inhibition of BCR signaling.
  • the invention relates to characterizing the anti-Fc ⁇ RIIB antibodies of the invention for Fc ⁇ RII-mediated signaling in human monocytes/macrophages.
  • Coaggregation of Fc ⁇ RIIB with a receptor bearing the immunoreceptor tyrosine-based activation motif (ITAM) acts to down-regulate Fc ⁇ R-mediated phagocytosis using SHIP as its effector (Tridandapani et al. 2002, J. Biol. Chem. 277(7):5082-9).
  • Coaggregation of Fc ⁇ RIIA with Fc ⁇ RIIB results in rapid phosphorylation of the tyrosine residue on Fc ⁇ RIIB 's ITIM motif, leading to an enhancement in phosphorylation of SHIP, association of SHIP with She, and phosphorylation of proteins having the molecular weight of 120 and 60-65 kDa.
  • coaggregation of Fc ⁇ RIIA with Fc ⁇ RIIB results in down-regulation of phosphorylation of Akt, which is a serine- threonine kinase that is involved in cellular regulation and serves to suppress apoptosis.
  • the invention further encompasses characterizing the anti-Fc ⁇ RIIB antibodies of the invention for their inhibition of Fc ⁇ R-mediated phagocytosis in human monocytes/macrophages.
  • THP-I can be stimulated either with Fab fragments of mouse monoclonal antibody IV.3 against Fc ⁇ RII and goat anti-mouse antibody (to aggregate Fc ⁇ RIIA alone), or with whole IV.3 mouse monoclonal antibody and goat anti-mouse antibody (to coaggregate Fc ⁇ RIIA and Fc ⁇ RIIB).
  • modulation of down stream signaling molecules such as tyrosine phosphorylation of Fc ⁇ RIIB, phosphorylation of SHIP, association of SHIP with She, phosphorylation of Akt, and phosphorylation of proteins having the molecular weight of 120 and 60-65 kDa can be assayed upon addition of antibodies of the invention to the stimulated cells.
  • Fc ⁇ RIIB-dependent phagocytic efficiency of the monocyte cell line can be directly measured in the presence and absence of the antibodies of the invention.
  • Another exemplary assay for determining inhibition of Fc ⁇ R-mediated phagocytosis in human monocytes/macrophages by the antibodies of the invention can comprise the following: stimulating THP-I cells with either Fab of IV.3 mouse anti-Fc ⁇ RII antibody and goat anti-mouse antibody (to aggregate Fc ⁇ RIIA alone and elicit Fc ⁇ RIIA-mediated signaling); or with mouse anti-Fc ⁇ RII antibody and goat anti-mouse antibody (to coaggregate Fc ⁇ RIIA and Fc ⁇ RIIB and inhibiting Fc ⁇ RIIA-mediated signaling.
  • Cells that have been stimulated with mouse anti-Fc ⁇ RII antibody and goat anti-mouse antibody can be further pre-incubated with the antibodies of the invention. Measuring Fc ⁇ RIIA-dependent activity of stimulated cells that have been pre-incubated with antibodies of the invention and cells that have not been pre-incubated
  • NYJD 16033542 with the antibodies of the invention and comparing levels of Fc ⁇ RIIA-dependent activity in these cells would indicate a modulation of Fc ⁇ RIIA-dependent activity by the antibodies of the invention.
  • the exemplary assay described can be used for example, to identify antibodies that block ligand binding of Fc ⁇ RIIB receptor and antagonize Fc ⁇ RIIB -mediated inhibition of Fc ⁇ RIIA signaling by preventing coaggregation of Fc ⁇ RIIB and Fc ⁇ RIIA.
  • This assay likewise identifies antibodies that enhance coaggregation of Fc ⁇ RIIB and Fc ⁇ RIIA and agonize Fc ⁇ RIIB - mediated inhibition of Fc ⁇ RIIA signaling.
  • the invention relates to characterizing the function of the antibodies of the invention by measuring the ability of THP-I cells to phagocytose fluoresceinated IgG-opsonized sheep red blood cells (SRBC) by methods previously described (Tridandapani et al, 2000, J. Biol. Chem. 275: 20480-7).
  • SRBC phagocytose fluoresceinated IgG-opsonized sheep red blood cells
  • an exemplary assay for measuring phagocytosis comprises of: treating THP-I cells with the antibodies of the invention or with a control antibody that does not bind to Fc ⁇ RII, comparing the activity levels of said cells, wherein a difference in the activities of the cells ⁇ e.g., rosetting activity (the number of THP-I cells binding IgG-coated SRBC), adherence activity (the total number of SRBC bound to THP-I cells), and phagocytic rate) would indicate a modulation of Fc ⁇ RIIA-dependent activity by the antibodies of the invention.
  • rosetting activity the number of THP-I cells binding IgG-coated SRBC
  • adherence activity the total number of SRBC bound to THP-I cells
  • phagocytic rate a modulation of Fc ⁇ RIIA-dependent activity by the antibodies of the invention.
  • This assay can be used to identify, for example, antibodies that block ligand binding of Fc ⁇ RIIB receptor and antagonize Fc ⁇ RIIB -mediated inhibition of phagocytosis. This assay can also identify antibodies that enhance Fc ⁇ RIIB -mediated inhibition of Fc ⁇ RIIA signaling.
  • the antibodies of the invention modulate Fc ⁇ RIIB - dependent activity in human monocytes/macrophages in at least one or more of the following ways: modulation of downstream signaling molecules ⁇ e.g., modulation of phosphorylation state of Fc ⁇ RIIB, modulation of SHIP phosphorylation, modulation of SHIP and She association, modulation of phosphorylation of Akt, modulation of phosphorylation of additional proteins around 120 and 60-65 kDa) and modulation of phagocytosis.
  • modulation of downstream signaling molecules ⁇ e.g., modulation of phosphorylation state of Fc ⁇ RIIB, modulation of SHIP phosphorylation, modulation of SHIP and She association, modulation of phosphorylation of Akt, modulation of phosphorylation of additional proteins around 120 and 60-65 kDa
  • modulation of phagocytosis modulation of downstream signaling molecules ⁇ e.g., modulation of phosphorylation state of Fc ⁇ RIIB, modul
  • the invention encompasses characterization of the antibodies of the invention using assays known to those skilled in the art for identifying the effect of the antibodies on effector cell function of therapeutic antibodies, e.g., their ability to enhance tumor- specific ADCC activity of therapeutic antibodies.
  • Therapeutic antibodies that may be used in accordance with the methods of the invention include but are not limited to anti-tumor antibodies, anti-viral antibodies, anti-microbial antibodies ⁇ e.g., bacterial and unicellular parasites), examples of which are disclosed herein (Section 5.3.4).
  • the invention encompasses
  • NYJD 16033542 characterizing the antibodies of the invention for their effect on Fc ⁇ R-mediated effector cell function of therapeutic antibodies, e.g., tumor-specific monoclonal antibodies.
  • effector cell functions include but are not limited to, antibody-dependent cell mediated cytotoxicity, phagocytosis, opsonization, opsonophagocytosis, CIq binding, and complement dependent cell mediated cytotoxicity. Any cell-based or cell free assay known to those skilled in the art for determining effector cell function activity can be used (For effector cell assays, see Perussia et al, 2000, Methods MoI. Biol.
  • Antibodies of the invention can be assayed for their effect on Fc ⁇ R-mediated
  • ADCC activity of therapeutic antibodies in effector cells e.g., natural killer cells, using any of the standard methods known to those skilled in the art (See e.g., Perussia et al., 2000, Methods MoI. Biol. 121: 179-92).
  • "Antibody-dependent cell-mediated cytotoxicity” and "ADCC” as used herein carry their ordinary and customary meaning in the art and refer to an in vitro cell- mediated reaction in which nonspecific cytotoxic cells that express Fc)Rs (e.g., monocytic cells such as Natural Killer (NK) cells and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • Fc cytotoxic cells that express Fc)Rs
  • NK Natural Killer
  • any effector cell with an activating Fc)R can be triggered to mediate ADCC.
  • the primary cells for mediating ADCC are NK cells which express only Fc)RIII, whereas monocytes, depending on their state of activation, localization, or differentiation, can express Fc)RI, Fc)RII, and Fc)RIII.
  • NK cells which express only Fc)RIII
  • monocytes depending on their state of activation, localization, or differentiation, can express Fc)RI, Fc)RII, and Fc)RIII.
  • Effector cells are leukocytes which express one or more Fc)Rs and perform effector functions.
  • the cells express at least Fc)RIII and perform ADCC effector function.
  • Effector cells that may be used in the methods of the invention include but are not limited to peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, and neutrophils; with PBMCs and NK cells being preferred.
  • PBMC peripheral blood mononuclear cells
  • NK natural killer cells
  • the effector cells may be isolated from a native source thereof, e.g., from blood or PBMCs as described herein.
  • the effector cells used in the ADCC assays of the invention are peripheral blood mononuclear cells (PBMC) that are preferably purified from normal human blood, using standard methods known to one
  • PBMC peripheral blood mononuclear cells
  • PBMCs may be isolated by layering whole blood onto Ficoll-Hypaque and spinning the cells at 500g, at room temperature for 30 minutes.
  • the leukocyte layer can be harvested as effector cells.
  • Other effector cells that may be used in the ADCC assays of the invention include but are not limited to monocyte-derived macrophages (MDMs).
  • MDMs that are used as effector cells in the methods of the invention are preferably obtained as frozen stocks or used fresh, (e.g., from Advanced Biotechnologies, MD).
  • elutriated human monocytes are used as effector cells in the methods of the invention.
  • Elutriated human monocytes express activating receptors, Fc ⁇ RIIIA and Fc ⁇ RIIA and the inhibitory receptor, Fc ⁇ RIIB.
  • Human monocytes are commercially available and may be obtained as frozen stocks, thawed in basal medium containing 10% human AB serum or in basal medium with human serum containing cytokines.
  • Levels of expression of Fc ⁇ Rs in the cells may be directly determined; e.g. using FACS analysis. Alternatively, cells may also be allowed to mature to macrophages in culture. The level of Fc ⁇ RIIB expression may be increased in macrophages.
  • Antibodies that may be used in determining the expression level of Fc ⁇ Rs include but are not limited to anti-human Fc ⁇ RIIA antibodies, e.g., IV.3-FITC; anti- Fc ⁇ RI antibodies, e.g., 32.2 FITC; and anti- Fc ⁇ RIIIA antibodies, e.g., 3G8-PE.
  • Target cells used in the ADCC assays of the invention include, but are not limited to, breast cancer cell lines, e.g., SK-BR-3 with ATCC accession number HTB-30 (see, e.g., Tremp et al, 1976, Cancer Res. 33-41); B-lymphocytes; cells derived from Burkitts lymphoma, e.g., Raji cells with ATCC accession number CCL-86 (see, e.g., Epstein et al, 1965, J. Natl. Cancer Inst. 34: 231-240), Daudi cells with ATCC accession number CCL-213 (see, e.g., Klein et al., 1968, Cancer Res.
  • breast cancer cell lines e.g., SK-BR-3 with ATCC accession number HTB-30 (see, e.g., Tremp et al, 1976, Cancer Res. 33-41); B-lymphocytes; cells derived from Burkitts lymphoma, e.g.
  • ovarian carcinoma cell lines e.g., OVCAR-3 with ATCC accession number HTB-161 (see, e.g., Hamilton, Young et al, 1983), SK-OV-3, PA-I, CAOV3, OV-90, and IGROV-I (available from the NCI repository Benard et al, 1985, Cancer Research, 45:4970-9; which is incorporated herein by reference in its entirety.
  • the target cells must be recognized by the antigen binding site of the antibody to be assayed.
  • the target cells for use in the methods of the invention may have low, medium, or high expression level of a cancer antigen.
  • the expression levels of the cancer antigen may be determined using common methods known to one skilled in the art, e.g., FACS analysis.
  • the invention encompasses the use of ovarian cancer cells such as IGROV-I, wherein Her2/neu is expressed at different levels, or OV-CAR-3 (ATCC Assession Number HTB-161; characterized by a lower expression of Her2/neu than SK-BR-3, the breast carcinoma cell line).
  • ovarian carcinoma cell lines that may be used as target cells in the methods of the invention include OVCAR-8
  • NYJD 1603354.2 (Hamilton et al., 1983, Cancer Res. 43:5379-89,which is incorporated herein by reference in its entirety); SK-OV-3 (ATCC Accession Number HTB-77); Caov-3 (ATCC Accession Number HTB-75); PA-I (ATCC Accession Number CRL- 1572); OV-90 (ATCC Accession Number CRL-11732); and OVCAR-4.
  • breast cancer cell lines that may be used in the methods of the invention include BT-549 (ATCC Accession Number HTB- 122), MCF7 (ATCC Accession Number HTB-22), and Hs578T (ATCC Accession Number HTB-126), all of which are available from the NCI repository and ATCC and incorporated herein by reference.
  • cell lines that may be used in the methods of the invention include but are not limited to CCRF-CEM (leukemia); HL-60 (TB, leukemia); MOLT-4 (leukemia); RPMI-8226 (leukemia); SR (leukemia); A549 (Non-small cell lung); EKVX (Non-small cell lung); HOP-62 (Non-small cell lung); HOP-92 (Non-small cell lung); NC1-H226 (Non-small cell lung); NC1-H23 (Non-small cell lung); NC1-H322M (Non-small cell lung); NC1-H460 (Non-small cell lung); NC1-H522 (Non-small cell lung); COLO 205 (Colon); HCC-2998 (Colon); HCT-116 (Colon); HCT-15 (Colon); HT29 (Colon); KM12 (Colon); SW-620 (Colon); SF-268 (CNS); SF-295 (CNS); SF- 5
  • An exemplary assay for determining the effect of the antibodies of the invention on the ADCC activity of therapeutic antibodies is based on a 51 Cr release assay comprising of: labeling target cells with [ 51 Cr]Na 2 CrO 4 (this cell-membrane permeable molecule is commonly used for labeling since it binds cytoplasmic proteins and although spontaneously released from the cells with slow kinetics, it is released massively following target cell lysis); preferably, the target cells express one or more tumor antigens, osponizing the target cells with one or more antibodies that immunospecifically bind the tumor antigens expressed on the cell surface of the target cells, in the presence and absence of an antibody of the invention, e.g., 2B6, 3H7, combining the opsonized radiolabeled target cells with effector cells in a microtitre plate at an appropriate ratio of target cells to effector cells; incubating the mixture of cells preferably for 16-18 hours, preferably at 37 0 C; collecting supernatants; and analyzing the
  • a graph can be generated by varying either the target: effector cell ratio or antibody concentration.
  • the antibodies of the invention are characterized for antibody dependent cellular cytotoxicity (ADCC) in accordance with the method described earlier, see, e.g., Ding et ah, Immunity, 1998, 8:403-11; which is incorporated herein by reference in its entirety.
  • ADCC antibody dependent cellular cytotoxicity
  • the invention encompasses characterizing the function of the antibodies of the invention in enhancing ADCC activity of therapeutic antibodies in an in vitro based assay and/or in an animal model.
  • the invention encompasses determining the function of the antibodies of the invention in enhancing tumor specific ADCC using an ovarian cancer model and/or breast cancer model.
  • the ADCC assays of the invention are done using more than one cancer cell line, characterized by the expression of at least one cancer antigen, wherein the expression level of the cancer antigen is varied among the cancer cell lines used.
  • performing ADCC assays in more than one cell line wherein the expression level of the cancer antigen is varied will allow determination of stringency of tumor clearance of the antibodies of the invention.
  • the ADCC assays of the invention are done using cancer cell lines with different levels of expression of a cancer antigen.
  • OVCAR3, an ovarian carcinoma cell line can serve as the tumor target expressing the tumor antigens, Her2/neu and TAG-72; human monocytes, that express the activating Fc ⁇ RIIIA and Fc ⁇ RIIA and inhibitory Fc ⁇ RIIB, can be used as effectors; and tumor specific murine antibodies, ch4D5 and chCC49, can be used as the tumor specific antibodies.
  • OVCAR-3 cells are available from ATCC (Accession Number HTB-161). Preferably, OVCAR-3 cells are propagated in medium supplemented with 0.01 mg/ml bovine insulin.
  • 5 x 10 6 viable OVCAR-3 cells may be injected subcutaneously (s.c) into age and weight matched nude athymic mice with Matrigel (Becton Dickinson).
  • the estimated weight of the tumor can be calculated by the formula: length-(width) 2 /2, and preferably does not exceed 3 grams.
  • Anchorage-dependent tumor can be isolated after 6-8 weeks, and the cells can be dissociated by adding 1 ⁇ g of Collagenase (Sigma) per gram of tumor and a 5 mg/mL RNase,
  • NYJD 1603354.2 passed through a cell strainer and nylon mesh to isolate cells. Cells can then be frozen for long- term storage for s.c. injection for establishment of the xenograft model.
  • Hybridomas secreting CC49 and 4D5 antibodies are available with ATCC
  • the 4D5 and CC49 antibodies are chimerized using standard methods known to one skilled in the art so that the human Fc sequence, e.g., human constant region of IgGl, is grafted onto the variable region of the murine antibodies in order to provide the effector function.
  • CC49 is directed to TAG-72; a high molecular weight mucin that is highly expressed on many adenocarcinoma cells and ovarian carcinoma (Lottich et al, 1985 Breast Cancer Res. Treat. 6(l):49-56; Mansi et al, 1989 Int. J. Rod. Appl. Instrum B. 16(2): 127-35; Colcher et al, 1991 Int. J. Rod. Appl Instrum B. 18:395- 41; all of which are incorporated herein by reference in their entirety).
  • 4D5 is directed to human epidermal growth factor receptor 2 (Carter et al, 1992, Proc. Natl. Acad. Sci. USA, 89: 4285-9 which is incorporated herein by reference).
  • Antibodies of the invention can then be utilized to investigate the enhancement of ADCC activity of the tumor specific antibodies, by blocking the inhibitory Fc ⁇ RIIB.
  • the inhibitory receptor e.g., Fc ⁇ RIIA
  • Fc ⁇ RIIB the expression of the inhibitory receptor (Fc ⁇ RIIB) is enhanced and this limits the clearance of tumors as the ADCC activity of Fc ⁇ RIIA is suppressed.
  • antibodies of the invention can serve as a blocking antibody, i.e., an antibody that will prevent the inhibitory signal from being activated and thus the activation signal, e.g., ADCC activity, will be sustained for a longer period and may result in potent tumor clearance.
  • a blocking antibody i.e., an antibody that will prevent the inhibitory signal from being activated and thus the activation signal, e.g., ADCC activity, will be sustained for a longer period and may result in potent tumor clearance.
  • the antibodies of the invention for use in enhancement of ADCC activity have been modified to comprise at least one amino acid modification, so that their binding to Fc ⁇ R has been diminished, most preferably abolished.
  • the antibodies of the invention have been modified to comprise at least one amino acid modification which reduces the binding of the constant domain to an activating Fc ⁇ R, e.g., Fc ⁇ RIILA, Fc ⁇ RIIA, as compared to a wild type antibody of the invention while retaining maximal Fc ⁇ RIIB blocking activity.
  • Antibodies of the invention may be modified in accordance with any method known to one skilled in the art or disclosed herein. Any amino acid modification which is
  • NYJD 1603354 2 known to disrupt effector function may be used in accordance with the methods of the invention such as those disclosed in U.S. Application serial Nos. 60/439,498 (filed January 9, 2003); and 60/456,041 (filed March 19, 2003); both of which are incorporated herein by reference in their entireties.
  • antibodies of the invention are modified so that position 265 is modified, e.g., position 265 is substituted with alanine.
  • the murine constant region of an antibody of the invention is swapped with the corresponding human constant region comprising a substitution of the amino acid at position 265 with alanine, so that the effector function is abolished while Fc)RIIB blocking activity is maintained.
  • antibodies of the invention are modified so that position 297 is modified, e.g., position 297 is substituted with glutamine, so that the N-linked glycosylation site is eliminated (see, e.g., Jefferies et al, 1995, Immunol, left 44:111-7;; Lund et al, 1996, 7.
  • the murine constant region of an antibody of the invention is swapped with the corresponding human constant region comprising a substitution of the amino acid at position 265 and/or 297, so that the effector function is abolished while Fc ⁇ RIIB blocking activity is maintained.
  • An exemplary assay for determining the ADCC activity of the tumor specific antibodies in the presence and absence of the antibodies of the invention is a non-radioactive europium based fluorescent assay (BATDA, Perkin Elmer) and may comprise the following: labeling the targets cells with an acteoxylmethyl ester of fluorescence-enhancing ester that forms a hydrophilic ligand (TDA) with the membrane of cells by hydrolysis of the esters; this complex is unable to leave the cell and is released only upon lysis of the cell by the effectors; adding the labeled targets to the effector cells in presence of anti-tumor antibodies and an antibody of the invention; incubating the mixture of the target and effector cells a for 6 to 16 hours, preferably at 37 0 C.
  • BATDA non-radioactive europium based fluorescent assay
  • ADCC activity can be assayed by measuring the amount of ligand that is released and interacts with europium (DELFIA reagent; PerkinElmer).
  • DELFIA reagent PerkinElmer
  • the ligand and the europium form a very stable and highly fluorescent chelate (EuTDA) and the measured fluorescence is directly proportional to the number of cells lysed. Percent specific lysis can be
  • NYJD 1603354.2 calculated using the formula: (Experimental lysis-antibody-independent lysis/maximal lysis antibody-independent lysis x 100%).
  • the invention encompasses radioactive-based ADCC assays, such as 51 Cr release assay. Radioactive-based assays may be done instead of or in combination with fluorescent-based ADCC assays.
  • An exemplary 51 Cr release assay for characterizing the antibodies of the invention can comprise the following: labeling 1-2 xlO 6 target cells such as OVCAR-3 cells with 51 Cr; opsonizing the target cells with antibodies 4D5 and CC49 in the presence and absence of an antibody of the invention and adding 5 x 10 3 cells to 96 well plate.
  • 4D5 and CC49 are at a concentration varying from 1-15 ⁇ g/mL; adding the opsonized target cells to monocyte- derived macrophages (MDM) (effector cells); preferably at a ratio varying from 10:1 to 100:1; incubating the mixture of cells for 16-18 hours at 37°C; collecting supernatants; and analyzing the radioactivity in the supernatant.
  • MDM monocyte- derived macrophages
  • the in vivo activity of the Fc ⁇ RIIB antibodies of the invention is determined in xenograft human tumor models.
  • Tumors may be established using any of the cancer cell lines described supra.
  • the tumors will be established with two cancer cell lines, wherein the first cancer cell line is characterized by a low expression of a cancer antigen and a second cancer cell line, wherein the second cancer cell line is characterized by a high expression of the same cancer antigen.
  • Tumor clearance may then be determined using methods known to one skilled in the art, using an anti-tumor antibody which immunospecifically binds the cancer antigen on the first and second cancer cell line, and an appropriate mouse model, e.g., a Balb/c nude mouse model (e.g., Jackson Laboratories, Taconic), with adoptively transferred human monocytes and MDMs as effector cells. Any of the antibodies described supra may then be tested in this animal model to evaluate the role of anti-Fc ⁇ RIIB antibody of the invention in tumor clearance.
  • an anti-tumor antibody which immunospecifically binds the cancer antigen on the first and second cancer cell line
  • an appropriate mouse model e.g., a Balb/c nude mouse model (e.g., Jackson Laboratories, Taconic), with adoptively transferred human monocytes and MDMs as effector cells.
  • Any of the antibodies described supra may then be tested in this animal model to evaluate the role of anti-Fc ⁇ RIIB antibody of the invention in tumor clearance.
  • mice that may be used in the invention include for example Fc ⁇ RIII -/- (where Fc ⁇ RIIIA is knocked out); Fc ⁇ -/- nude mice (where Fc ⁇ RI and Fc ⁇ RIIIA are knocked out); or human Fc ⁇ RIIB knock in mice or a transgenic knock-in mice, where mouse/cgr2 and fcgr3 loci on chromosome 1 are inactivated and the mice
  • NYJD 16033542 express human Fc)RIIA, human Fc ⁇ RIIA human Fc ⁇ RIIB, human FcyRIIC, human Fc)RIIIA, and human Fc)RIIIB.
  • An exemplary method for testing the in vivo activity of an antibody of the invention may comprise the following: establishing a xenograft murine model using a cancer cell line characterized by the expression of a cancer antigen and determining the effect of an antibody of the invention on an antibody specific for the cancer antigen expressed in the cancer cell line in mediating tumor clearance.
  • the in vivo activity is tested parallel using two cancer cell lines, wherein the first cancer cell line is characterized by a first cancer antigen expressed at low levels and a second cancer cell line, characterized by the same cancer antigen expressed at a higher level relative to the first cancer cell line.
  • tumors may be established with the IGROV-I cell line and the effect of an anti-FcyRIIB antibody of the invention in tumor clearance of a Her2/neu specific antibody may be assessed.
  • 5xlO 6 viable cells e.g., IGROV-I, SKBR3
  • mice e.g., 8 age and weight matched femal nude athymic mice using for example Matrigel (Becton Dickinson).
  • the estimated weight of the tumor may be determined by the formula: length x (width) 2 /2; and preferably does not exceed 3 grams.
  • Injection of IGROV-I cells s.c.
  • the IGROV-I cells form tumors within 5 weeks, at day 1 after tumor cell injection, monocytes as effectors are co-injected i.p. along with a therapeutic antibody specific for Her2/neu, e.g., Ch4D5, and an antibody of the invention; e.g. chimeric 2B6 or 3H7 as described supra.
  • the antibodies are injected at 4 ⁇ g each per gram of mouse body weight (mbw).
  • mice will receive no therapeutic antibody but will be injected with a chimeric 4D5 comprising a N297A mutation and human IgGl as isotype control antibodies for the anti-tumor and anti- Fc ⁇ RIIB antibodies, respectively. Mice may be placed in groups of 4 and monitored three times weekly.
  • Table 3 below is an exemplary setup for tumor clearance studies in accordance with the invention. As shown in Table 3, six groups of 8 mice each will be needed for testing the role of an antibody of the invention in tumor clearance, wherein one target and effector cell combination is used and wherein two different combinations of the antibody concentration are
  • NYJD 1603354.2 used.
  • group A only tumor cells are injected; in group B tumor cells and monocytes are injected; in group C, tumor cells, monocytes, an anti-tumor antibody (ch4D5) are injected; in group D, tumor cells, monocytes, anti-tumor antibody, and an anti-Fc ⁇ RII antibody are injected; in group E, tumor cells, monocytes and an anti-Fc ⁇ RIIB antibody are injected; in group F, tumor cells, monocytes, Ch4D5 (N297Q), and human IgGl are injected.
  • group A tumor cells are injected in group B tumor cells and monocytes are injected; in group C, tumor cells, monocytes, an anti-tumor antibody (ch4D5) are injected; in group D, tumor cells, monocytes, anti-tumor antibody, and an anti-Fc ⁇ RII antibody are injected; in group E, tumor cells, monocytes and an anti-Fc ⁇ RIIB antibody are injected;
  • the endpoint of the xenograft tumor models is determined based on the size of the tumors, weight of mice, survival time and histochemical and histopathological examination of the cancer, using methods known to one skilled in the art.
  • Each of the groups of mice in Table 3 will be evaluated. Mice are preferably monitored three times a week. Criteria for tumor growth may be abdominal distention, presence of palpable mass in the peritoneal cavity. Preferably estimates of tumor weight versus days after inoculation will be calculated. A comparison of the aforementioned criteria of mice in Group D compared to those in other groups will define the role of an antibody of the invention in enhancement of tumor clearance. Preferably, antibody-treated animals will be under observation for an additional 2 months after the control group.
  • human Fc ⁇ RIIB "knock in” mice expressing human Fc ⁇ RIIB
  • Fc ⁇ RIIB on murine effector cells may be used in establishing the in vivo activity of the antibodies of the invention, rather than adoptively transferring effector cells.
  • Founder mice expressing the human Fc ⁇ RIIB may be generated by "knocking in” the human Fc ⁇ RIIB onto the mouse Fc ⁇ RIIB locus. The founders can then be back-crossed onto the nude background and
  • NYJD: 16033542 will express the human Fc ⁇ RIIB receptor.
  • the resulting murine effector cells will express endogenous activating Fc ⁇ RI and Fc ⁇ RIIIA and inhibitory human Fc ⁇ RIIB receptors.
  • the in vivo activity of the antibodies of the invention may be further tested in a xenograft murine model with human primary tumor derived cells, such as human primary ovarian and breast carcinoma derived cells. Ascites and pleural effusion samples from cancer patients may be tested for expression of Her2/neu, using methods known to one skilled in the art.
  • Samples from ovarian carcinoma patients may be processed by spinning down the ascites at 637Og for 20 minutes at 4°C, lysing the red blood cells, and washing the cells with PBS. Once the expression of Her2/neu in tumor cells is determined, two samples, a median and a high expressor may be selected for s.c. inoculation to establish the xenograft tumor model. The isolated tumor cells will then be injected i.p. into mice to expand the cells. Approximately 10 mice may be injected i.p. and each mouse ascites further passaged into two mice to obtain ascites from a total of 20 mice which can be used to inject a group of 80 mice. Pleural effusion samples may be processed using a similar method as ascites.
  • the Her2/neu+ tumor cells from pleural effusion samples may be injected into the upper right & left mammary pads of the mice.
  • the neoplastic cells may be expanded in vitro.
  • tumor cells may be purified using CC49 antibody (anti-TAG-72)-coated magnetic beads as described previously, see, e.g., Barker et al., 2001, Gynecol. Oncol. 82:57-63, which is incorporated herein by reference in its entirety.
  • magnetic beads coated with CC49 antibody can be used to separate the ovarian tumor cells that will be detached from the beads by an overnight incubation at 37°C.
  • negative depletion using a cocktail of antibodies such as those provided by Stem Cell Technologies, Inc., Canada, may be used to enrich the tumor cells.
  • tumors markers besides Her2/neu may be used to separate tumor cells obtained from the ascites and pleural effusion samples from non- tumor cells.
  • CD44 an adhesion molecule
  • B38.1 a breast/ovarian cancer-specific marker
  • CD24 an adhesion molecule
  • immunohistochemistry and histochemistry is performed on ascites and pleural effusion of patients to analyze structural characteristics of the neoplasia.
  • NYJD 16033542 methods are known to one skilled in the art and encompassed within the invention.
  • the markers that may be monitored include for example cytokeratin (to identify ovarian neoplastic and mesothelial cells from inflammatory and mesenchymal cells); calretinin (to separate mesothelial from Her2neu positive neoplastic cells); and CD45 (to separate inflammatory cells from the rest of the cell population in the samples). Additional markers that may be followed include CD3 (T cells), CD20 (B cells), CD56 (NK cells), and CD 14 (monocytes). It will be appreciated by one skilled in the art that the immunohistochemistry and histochemistry methods described supra, are analogously applied to any tumor cell for use in the methods of the invention.
  • tumors are established using carcinoma cell lines such as IGROV-I, OVCAR-8, SK-B, and OVCAR-3 cells and human ovarian carcinoma ascites and pleural effusion from breast cancer patients.
  • carcinoma cell lines such as IGROV-I, OVCAR-8, SK-B, and OVCAR-3 cells and human ovarian carcinoma ascites and pleural effusion from breast cancer patients.
  • the ascites preferably contain both the effectors and the tumor targets for the antibodies being tested. Human monocytes will be transferred as effectors.
  • the in vivo activity of the antibodies of the invention may also be tested in an animal model, e.g., Balb/c nude mice, injected with cells expressing Fc ⁇ RIIB, including but not limited to SK-BR-3 with ATCC accession number HTB-30 ⁇ see, e.g., Tremp et al, 1976, Cancer Res. 33-41); B-lymphocytes; cells derived from Burkitts lymphoma, e.g., Raji cells with ATCC accession number CCL-86 ⁇ see, e.g., Epstein et al, 1965, /. Natl. Cancer Inst.
  • an animal model e.g., Balb/c nude mice, injected with cells expressing Fc ⁇ RIIB, including but not limited to SK-BR-3 with ATCC accession number HTB-30 ⁇ see, e.g., Tremp et al, 1976, Cancer Res. 33-41); B-lymphocytes; cells derived from Burkitts
  • OVCAR-3 with ATCC accession number HTB-161 see, e.g., Hamilton, Young et al, 1983
  • SK-OV-3, PA-I, CAOV3, OV-90, and IGROV-I available from the NCI repository Benard et al, 1985, Cancer Research, 45:4970-9; which is incorporated herein by reference in its entirety.
  • An exemplary assay for measuring the in vivo activity of the antibodies of the invention may comprise the following: Balb/c Nude female mice (Taconic, MD) are injected at day 0 with cells expressing Fc ⁇ RIIB such as 5xlO 6 Daudi cells for example by the subcutaneous route. Mice ⁇ e.g., 5 mice per group) also receive i.p. injection of PBS (negative control), ch 4.4.20 (anti-FITC antibody) as a negative control, and as a positive control another therapeutic cancer antibody such as those disclosed herein, e.g., Rituxan, ⁇ e.g., at 10 ⁇ g/g) or 10 ⁇ g/g ch2B6 once a week starting at day 0. Mice are observed, e.g., twice a week following injection, and
  • tumor size (length and width) is determined using for example a caliper. Tumor weight in mg is estimated using the formula: (length x width 2 )/2.
  • the antibodies of the invention have an enhanced efficacy in decreasing tumor relative to a cancer therapeutic antibody when administered at the same dose, e.g., 10 ⁇ g /g, over a time period of at least 14 days, at least 21 days, at least 28 days, or at least 35 days.
  • the antibodies of the invention reduce tumor size by at least 10 fold, at least 100 fold, at least 1000 fold relative to administration of a cancer therapeutic antibody at the same dose.
  • the antibodies of the invention completely abolish the tumor.
  • the present invention also includes polynucleotides that encode the antibodies of the invention (e.g., mouse monoclonal antibody produced from clone 2B6 or 3H7, with ATCC accession numbers PTA-4591 and PTA-4592, respectively), or other monoclonal antibodies produced by immunization methods of the invention, and humanized versions thereof, and methods for producing same.
  • polynucleotides that encode the antibodies of the invention e.g., mouse monoclonal antibody produced from clone 2B6 or 3H7, with ATCC accession numbers PTA-4591 and PTA-4592, respectively
  • other monoclonal antibodies produced by immunization methods of the invention and humanized versions thereof, and methods for producing same.
  • the present invention encompass the polynucleotide encoding the heavy chain of the 2B6 antibody, with ATCC accession number PTA-4591.
  • the present invention also encompasses the polynucleotide encoding the light chain of the 2B6 antibody with ATCC accession number PTA-4591.
  • the methods of the invention also encompass polynucleotides that hybridize under various stringency, e.g., high stringency, intermediate or lower stringency conditions, to polynucleotides that encode an antibody of the invention.
  • the hybridization can be performed under various conditions of stringency.
  • procedures using conditions of low stringency are as follows (see also Shilo and Weinberg, 1981, Proc. Natl. Acad. ScL U.S.A. 78, 6789-6792).
  • Filters containing DNA are pretreated for 6 h at 40°C in a solution containing 35% formamide, 5X SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.1% PVP, 0.1% Ficoll, 1% BSA, and 500 ⁇ g/ml denatured salmon sperm DNA.
  • Hybridizations are carried out in the same solution with the following modifications: 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 ⁇ g/ml salmon sperm DNA, 10% (wt/vol) dextran sulfate, and 5-20 X 10 6 cpm 32 P-labeled probe is used.
  • Filters are incubated in hybridization mixture for 18-20 h at 4O 0 C, and then washed for 1.5 h at 55 0 C in a solution containing 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1% SDS. The wash solution is replaced with fresh solution and incubated an additional 1.5 h at 60°C. Filters are blotted dry and exposed for autoradiography. If
  • filters are washed for a third time at 65-68°C and re-exposed to film.
  • Other conditions of low stringency which may be used are well known in the art (e.g., as employed for cross-species hybridizations).
  • procedures using conditions of high stringency are as follows. Prehybridization of filters containing DNA is carried out for 8 h to overnight at 65 0 C in buffer composed of 6X SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 ⁇ g/ml denatured salmon sperm DNA.
  • Filters are hybridized for 48 h at 65 0 C in prehybridization mixture containing 100 ⁇ g/ml denatured salmon sperm DNA and 5-20 X 10 6 cpm of 32 P-labeled probe. Washing of filters is done at 37°C for 1 h in a solution containing 2X SSC, 0.01% PVP, 0.01% Ficoll, and 0.01% BSA. This is followed by a wash in 0.1X SSC at 50 0 C for 45 min before autoradiography. Other conditions of high stringency which may be used are well known in the art.
  • the polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art.
  • a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source (e.g., a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody of the invention, e.g., 2B6 or 3H7) by hybridization with Ig specific probes and/or PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art.
  • a suitable source e.g., a cDNA library generated from, or nucleic acid, preferably poly
  • nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et ⁇ l, 1990,
  • one or more of the CDRs are inserted within framework regions using routine recombinant DNA techniques.
  • the framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions ⁇ see, e.g., Chothia et al, 1998, J. MoI. Biol. 278: 457-479 for a listing of human framework regions).
  • the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds to Fc ⁇ RIIB with greater affinity than said antibody binds Fc ⁇ RIIA.
  • one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibodies of the invention to Fc ⁇ RIIB.
  • human libraries or any other libraries available in the art can be screened by standard techniques known in the art, to clone the nucleic acids encoding the antibodies of the invention.
  • the vector for the production of the antibody may be produced by recombinant DNA technology using techniques well known in the art. Methods which are well known to those skilled in the art can be used to construct expression vectors containing the antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. ⁇ See, for example, the techniques described in Sambrook et al., 1990,
  • An expression vector comprising the nucleotide sequence of an antibody can be transferred to a host cell by conventional techniques (e.g., electroporation, liposomal transfection, and calcium phosphate precipitation) and the transfected cells are then cultured by conventional techniques to produce the antibody of the invention.
  • the expression of the antibody is regulated by a constitutive, an inducible or a tissue, specific promoter.
  • the host cells used to express the recombinant antibodies of the invention may be either bacterial cells such as Escherichia coli, or, preferably, eukaryotic cells, especially for the expression of whole recombinant immunoglobulin molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for immunoglobulins (Foecking et al, 1998, Gene 45:101; Cockett et al, 1990, Bio/Technology 8:2).
  • host-expression vector systems may be utilized to express the antibodies of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of the antibodies may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express the antibodies of the invention in situ.
  • These include, but are not limited to, microorganisms such as bacteria (e.g., E. coli and B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing immunoglobulin coding sequences; yeast (e.g., Saccharomyces Pichi ⁇ ) transformed with recombinant yeast expression vectors containing immunoglobulin coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the immunoglobulin coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus (CaMV) and tobacco mosaic virus (TMV)) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing immunoglobulin coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 293T, 3T3 cells, lymphotic cells (see U.S.
  • mammalian cell systems e.g., COS,
  • Per C.6 cells rat retinal cells developed by Crucell harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
  • promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al, 1983, EMBO J.
  • pG ⁇ X vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a matrix glutathione- agarose beads followed by elution in the presence of free gluta-thione.
  • the pG ⁇ X vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • AcNPV Autographa californica nuclear polyhedrosis virus
  • the virus grows in Spodopteva frugiperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (e.g., the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (e.g., the polyhedrin promoter).
  • a number of viral-based expression systems may be utilized.
  • the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
  • Insertion in a non-essential region of the viral genome will result in a recombinant virus that is viable and capable of expressing the immunoglobulin molecule in infected hosts, (e.g., see Logan & Shenk, 1984, Proc. Natl. Acad. ScL USA 81:355-359).
  • Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert.
  • These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the
  • NYJD 1603354.2 inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et ai, 1987, Methods in Enzymol. 153:51-544).
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • Such mammalian host cells include but are not limited to CHO, VERY, BHK, HeIa, COS, MDCK, 293, 293T, 3T3, WI38, BT483, Hs578T, HTB2, BT20 and T47D, CRL7030 and Hs578Bst.
  • stable expression is preferred.
  • cell lines which stably express an antibody of the invention may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines which express the antibodies of the invention.
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibodies of the invention.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al, 1977, Cell 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1992, Proc. Natl. Acad. ScL USA 48:202), and adenine phosphoribosyltransferase (Lowy et al, 1980, Cell 22:817) genes can be employed in tk-, hgprt- or aprt- cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al, 1980, Proc. Natl. Acad Sci. USA 77:357; O'Hare et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc.
  • the expression levels of an antibody of the invention can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • vector amplification for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • a marker in the vector system expressing an antibody is amplifiable
  • increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the nucleotide sequence of the antibody, production of the antibody will also increase (Crouse et al, 1983, MoI Cell. Biol. 3:257).
  • the host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, 1986, Nature 322:52; Kohler, 1980, Proc. Natl. Acad. ScL USA 77:2197).
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
  • the antibody of the invention may be purified by any method known in the art for purification of an antibody, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • centrifugation e.g., centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • NYJD 16033542 5.3 PROPHYLACTICANDTHERAPEUTICMETHODS
  • the present invention encompasses antibody-based therapies which involve administering one or more of the antibodies of the invention to an animal, preferably a mammal, and most preferably a human, for preventing, treating, or ameliorating symptoms associated with a disease, disorder, or infection, associated with aberrant levels or activity of Fc ⁇ RIIB and/or treatable by altering immune function associated with Fc ⁇ RIIB activity or enhancing cytotoxic activity of a second therapeutic antibody or enhancing efficacy of a vaccine composition or breaking tolerance to an antigen.
  • therapy by administration of one or more antibodies of the invention is combine with administration of one or more therapies such as, but not limited to, chemotherapies, radiation therapies, hormonal therapies, and/or biological therapies/immunotherapies
  • Prophylactic and therapeutic compounds of the invention include, but are not limited to, proteinaceous molecules, including, but not limited to, peptides, polypeptides, proteins, including post-translationally modified proteins, antibodies, etc.; small molecules (less than 1000 daltons), inorganic or organic compounds; nucleic acid molecules including, but not limited to, double-stranded or single-stranded DNA, double-stranded or single-stranded RNA, as well as triple helix nucleic acid molecules.
  • Prophylactic and therapeutic compounds can be derived from any known organism (including, but not limited to, animals, plants, bacteria, fungi, and protista, or viruses) or from a library of synthetic molecules.
  • Antibodies may be provided in pharmaceutically acceptable compositions as known in the art or as described herein. As detailed below, the antibodies of the invention can be used in methods of treating cancer (particularly to enhance passive immunotherapy or efficacy of a cancer vaccine) or allergies (e.g., to enhance efficacy of a vaccine for treatment of allergy).
  • Antibodies of the present invention that function as a prophylactic and or therapeutic agent of a disease, disorder, or infection can be administered to an animal, preferably a mammal and most preferably a human, to treat, prevent or ameliorate one or more symptoms associated with the disease, disorder, or infection.
  • Antibodies of the invention can be administered in combination with one or more other prophylactic and/or therapeutic agents useful in the treatment, prevention or management of a disease, disorder, or infection associated with aberrant levels or activity of Fc ⁇ RIIB and/or treatable by altering immune function associated with Fc ⁇ RIIB activity.
  • one or more antibodies of the invention are administered to a mammal, preferably a human, concurrently with one or more
  • NYJD 1603354 2 other therapeutic agents useful for the treatment of cancer are not limited to the administration of prophylactic or therapeutic agents at exactly the same time, but rather it is meant that antibodies of the invention and the other agent are administered to a subject in a sequence and within a time interval such that the antibodies of the invention can act together with the other agent to provide an increased benefit than if they were administered otherwise.
  • each prophylactic or therapeutic agent may be administered at the same time or sequentially in any order at different points in time; however, if not administered at the same time, they should be administered sufficiently close in time so as to provide the desired therapeutic or prophylactic effect.
  • Each therapeutic agent can be administered separately, in any appropriate form and by any suitable route.
  • the prophylactic or therapeutic agents are administered less than 1 hour apart, at about 1 hour apart, at about 1 hour to about 2 hours apart, at about 2 hours to about 3 hours apart, at about 3 hours to about 4 hours apart, at about 4 hours to about 5 hours apart, at about 5 hours to about 6 hours apart, at about 6 hours to about 7 hours apart, at about 7 hours to about 8 hours apart, at about 8 hours to about 9 hours apart, at about 9 hours to about 10 hours apart, at about 10 hours to about 11 hours apart, at about 11 hours to about 12 hours apart, no more than 24 hours apart or no more than 48 hours apart.
  • two or more components are administered within the same patient visit.
  • the dosage amounts and frequencies of administration provided herein are encompassed by the terms therapeutically effective and prophylactically effective.
  • the dosage and frequency further will typically vary according to factors specific for each patient depending on the specific therapeutic or prophylactic agents administered, the severity and type of cancer, the route of administration, as well as age, body weight, response, and the past medical history of the patient. Suitable regimens can be selected by one skilled in the art by considering such factors and by following, for example, dosages reported in the literature and recommended in the Physician's Desk Reference (56 th ed., 2002).
  • the antibodies of this invention may also be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), which, for example, serve to increase the number or activity of effector cells which interact with the antibodies and, increase immune response.
  • lymphokines or hematopoietic growth factors such as, e.g., IL-2, IL-3 and IL-7
  • the antibodies of this invention may also be advantageously utilized in combination with one or more drugs used to treat a disease, disorder, or infection such as, for example anti-cancer agents or anti-viral agents, e.g., as detailed in sections 5.3.4 and 5.3.5 below.
  • Antibodies of the invention can be used alone or in combination with other therapeutic antibodies known in the art to prevent, inhibit or reduce the growth of primary tumores or metastasis of cancerous cells.
  • antibodies of the invention can be used in combination with antibodies used in cancer immunotherapy.
  • the invention encompasses the use of the antibodies of the invention in combination with another therapeutic antibody to enhance the efficacy of such immunotherapy by increasing the potency of the therapeutic antibody's effector function, e.g., ADCC, CDC, phagocytosis, opsonization, etc.
  • antibodies of the invention block Fc ⁇ RIIB, preferably on monocytes and macrophages and thus enhance the therapeutic benefits a clinical efficacy of tumor specific antibodies by, for example, enhancing clearance of the tumors mediated by activating fc ⁇ Rs.
  • the invention provides methods of preventing or treating cancer characterized by a cancer antigen, when administered in combination with another antibody that specifically binds a cancer antigen and is cytotoxic.
  • the antibodies of the invention are useful for prevention or treatment of cancer, particularly in potentiating the cytotoxic activity of cancer antigen-specific therapeutic antibodies with cytotoxic activity to enhance tumor cell killing by the antibodies of the invention and/or enhancing for example, ADCC activity or CDC activity of the therapeutic antibodies.
  • an antibody of the invention when administered alone or in combination with a cytotoxic therapeutic antibody, inhibits or reduces the growth of primary tumor or metastasis of cancerous cells by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to the growth of primary tumor or metastasis in absence of said antibody of the invention.
  • antibodies of the invention in combination with a cytotoxic therapeutic antibody inhibit or reduce the growth of primary tumor or metastasis of cancer by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to the growth or metastasis in absence of said antibodies.
  • the transition from a normal to a malignant state is a multistep process involving genetic and epigenetic changes.
  • CSF-I colony stimulating factor 1 or macrophage colony stimulating factor
  • angiogenic factors e.g., thymidine phosphorylase, vascular endothelial-derived growth factor; secretion of growth factors such as epidermal growth factor that could act as a paracrine factor on tumor cells, and thus promoting tumor cell migration and invasion into blood vessels.
  • the invention encompasses using the antibodies of the invention to block macrophage mediated tumor cell progression and metastasis.
  • the antibodies of the invention are particularly useful in the treatment of solid tumors, where macrophage infiltration occurs.
  • the antagonistic antibodies of the invention are particularly useful for controlling, e.g., reducing or eliminating, tumor cell metastasis, by reducing or eliminating the population of macrophages that are localized at the tumor site.
  • the antibodies of the invention are used alone to control tumor cell metastasis.
  • the antagonistic antibodies of the invention when administered alone bind the inhibitory Fc ⁇ RIIB on macrophages and effectively reduce the population of macrophages and thus restrict tumor cell progression.
  • the antagonistic antibodies of the invention reduce, or preferably eliminate macrophages that are localized at the tumor site, since Fc ⁇ RIIB is preferentially expressed on activated monocytes and macrophages including tumor-infiltrating macrophages.
  • the antibodies of the invention are used in the treatment of cancers that are characterized by the overexpression of CSF-I, including but not limited to breast, uterine, and ovarian cancers.
  • the invention further encompasses antibodies that effectively deplete or eliminate immune cells other than macrophages that express Fc ⁇ RIIB, e.g., dendritic cells and B- cells.
  • Effective depletion or elimination of immune cells using the antibodies of the invention may range from a reduction in population of the immune cells by 50%, 60%, 70%, 80%, preferably 90%, and most preferably 99%.
  • the antibodies of the invention have enhanced therapeutic efficacy either alone or in combination with a second antibody, e.g., a therapeutic antibody such as anti-tumor antibodes, anti-viral antibodies, and anti-microbial antibodies.
  • a therapeutic antibody such as anti-tumor antibodes, anti-viral antibodies, and anti-microbial antibodies.
  • the therapeutic antibodies have specificity for a cancer cell or an inflammatory cell.
  • the second antibody binds a normal cell.
  • the antibodies of the invention when used alone to deplete Fc ⁇ RIIB -expressing immune cells, the population of cells is redistributed so that effectively the cells that are remaining have the activating Fc receptors and thus the suppression by Fc)RIIB is alleviated.
  • a second antibody e.g., a therapeutic antibody the efficacy of the second antibody is enhanced by increasing the Fc-mediated effector function of the antibody.
  • liquid cancers cancers of the bone marrow, such as leukemias.
  • Solid cancers generally refer to cancers of organs and/or tissues.
  • Cancers and related disorders that can be treated or prevented by methods and compositions of the present invention include, but are not limited to, the following: Leukemias including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome, chronic leukemias such as but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, hairy cell leukemia; polycythemia vera; lymphomas such as but not limited to Hodgkin's disease, non- Hodgkin's disease; multiple myelomas such as but not limited to smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary
  • NYJD 1603354 2 adrenocortical carcinoma
  • thyroid cancer such as but not limited to papillary or follicular thyroid cancer, medullary thyroid cancer and anaplastic thyroid cancer
  • pancreatic cancer including but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor
  • pituitary cancers including but not limited to, Cushing's disease, prolactin-secreting tumor, acromegaly, and diabetes insipius
  • eye cancers including but not limited to, ocular melanoma such as iris melanoma, choroidal melanoma, and cilliary body melanoma, and retinoblastoma
  • vaginal cancers including but not limited to, squamous cell carcinoma, adenocarcinoma, and melanoma
  • vulvar cancer including but not limited to,
  • NYJD 1603354.2 cell carcinoma, squamous cell cancer, adenocarcinoma, carcinosarcoma.
  • cancers include myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangioendotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma and papillary adenocarcinomas (for a review of such disorders, see Fishman et al, 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al, 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America).
  • carcinoma including that of the bladder, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid and skin; including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Berketts lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyoscarcoma; other tumors, including melanoma, seminoma, tetratocarcinoma
  • cancers caused by aberrations in apoptosis would also be treated by the methods and compositions of the invention.
  • Such cancers may include but not be limited to follicular lymphomas, carcinomas with p53 mutations, hormone dependent tumors of the breast, prostate and ovary, and precancerous lesions such as familial adenomatous polyposis, and myelodysplastic syndromes.
  • malignancy or dysproliferative changes (such as metaplasias and dysplasias), or hyperproliferative disorders, are treated or prevented by the methods and compositions of the invention in the ovary, bladder, breast, colon, lung, skin, pancreas, or uterus.
  • sarcoma, melanoma, or leukemia is treated or prevented by the methods and compositions of the invention.
  • Cancers associated with the cancer antigens may be treated or prevented by administration of the antibodies of the invention in combination with an antibody that binds the cancer antigen and is cytotoxic.
  • the antibodies of the invention may be administered to the cancer antigens in combination with an antibody that binds the cancer antigen and is cytotoxic.
  • the antibodies of the invention may be administered to the cancer antigens in combination with an antibody that binds the cancer antigen and is cytotoxic.
  • the antibodies of the invention may be treated or prevented by administration of the antibodies of the invention in combination with an antibody that binds the cancer antigen and is cytotoxic.
  • the antibodies of the invention may be treated or prevented by administration of the antibodies of the invention in combination with an antibody that binds the cancer antigen and is cytotoxic.
  • NYJD: 1603354.2 enhance the antibody mediated cytotoxic effect of the antibody directed at the particular cancer antigen.
  • cancers associated with the following cancer antigen may be treated or prevented by the methods and compositions of the invention.
  • KS 1/4 pan-carcinoma antigen Perez and Walker, 1990, /. Immunol. 142:32-37; Bumal, 1988, Hybridoma 7(4):407-415
  • ovarian carcinoma antigen CA125
  • prostatic acid phosphate Tiailor et al, 1990, Nucl. Acids Res.
  • prostate specific antigen Henttu and Vihko, 1989, Biochem. Biophys. Res. Comm. 10(2):903-910; Israeli et al, 1993, Cancer Res. 53:227-230
  • melanoma-associated antigen p97 Estin et al, 1989, J. Natl. Cancer Instit. 81(6):445-44
  • melanoma antigen gp75 Vijayasardahl et al, 1990, J. Exp. Med.
  • HMW- MAA high molecular weight melanoma antigen
  • CEA carcinoembryonic antigen
  • TAG-72 Yokata et al, 1992, Cancer Res.
  • melanoma specific antigens such as ganglioside GD2 (Saleh et al, 1993, J.Immunol., 151, 3390-3398), ganglioside GD3 (Shitara et al, 1993, Cancer Immunol. Immunother. 36:373-380), ganglioside GM2 (Livingston et al, 1994, 7. CHn. Oncol. 12:1036- 1044), ganglioside GM3 (Hoon et al, 1993, Cancer Res.
  • tumor-specific transplantation type of cell-surface antigen such as virally-induced tumor antigens including T-antigen DNA tumor viruses and envelope antigens of RNA tumor viruses, oncofetal antigen-alpha-fetoprotein such as CEA of colon, bladder tumor oncofetal antigen (Hellstrom et al, 1985, Cancer. Res. 45:2210-2188), differentiation antigen such as human lung carcinoma antigen L6, L20 (Hellstrom et al, 1986, Cancer Res. 46:3917-3923), antigens of fibrosarcoma, human leukemia T cell antigen-Gp37 (Bhattacharya-Chatterjee et al, 1988, /. oflmmun.
  • TSTA tumor-specific transplantation type of cell-surface antigen
  • virally-induced tumor antigens including T-antigen DNA tumor viruses and envelope antigens of RNA tumor viruses
  • oncofetal antigen-alpha-fetoprotein such as CEA of colon
  • bladder tumor oncofetal antigen
  • neoglycoprotein neoglycoprotein
  • sphingolipids breast cancer antigen such as EGFR (Epidermal growth factor receptor), HER2 antigen (pl85 HER2 ), polymorphic epithelial mucin (PEM) (Hilkens et al, 1992, Trends in Bio. Chem.
  • NYJD 1603354.2 found in myeloid cells, VEP8, VEP9, MyI, VIM-D5,and 0 ⁇ 6-22 found in colorectal cancer, TRA-1-85 (blood group H), C14 found in colonic adenocarcinoma, F3 found in lung adenocarcinoma, AH6 found in gastric cancer, Y hapten, Le y found in embryonal carcinoma cells, TL5 (blood group A), EGF receptor found in A431 cells , E 1 series (blood group B) found in pancreatic cancer, FC 10.2 found in embryonal carcinoma cells, gastric adenocarcinoma, CO- 514 (blood group Le a ) found in adenocarcinoma, NS-IO found in adenocarcinomas, CO-43 (blood group Le b ), G49, EGF receptor, (blood group ALe b /Le y ) found in colonic adenocarcinoma, 19.9 found in colon cancer, gas
  • the antibodies of the invention can be used in combination with any therapeutic cancer antibodies known in the art to enhance the efficacy of treatment.
  • the antibodies of the invention can be used with any of the antibodies in Table 4, that have demonstrated therapeutic utility in cancer treatment.
  • the antibodies of the invention enhance the efficacy of treatment of the therapeutic cancer antibodies by enhancing at least one antibody- mediated effector function of said therapeutic cancer antibodies.
  • the antibodies enhance the efficacy of treatment by enhancing the complement dependent cascade of said therapeutic cancer antibodies.
  • the antibodies of the invention enhance the efficacy of treatment by enhancing the phagocytosis and opsonization of the targeted tumor cells.
  • the antibodies of the invention enhance the efficacy of treatment by enhancing antibody-dependent cell-mediated cytotoxicity ("ADCC”) in destruction of the targeted tumor cells.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Antibodies of the invention can also be used in combination with cytosine- guanine dinucleotides (“CpG”)-based products that have been developed (Coley Pharmaceuticals) or are currently being developed as activators of innate and acquired immune responses.
  • CpG cytosine- guanine dinucleotides
  • the invention encompasses the use of CpG 7909, CpG 8916, CpG 8954 (Coley Pharmaceuticals) in the methods and compositions of the invention for the treatment and/or prevention of cancer ⁇ See also Warren et al, 2002, Semin Oncol., 29(1 Suppl 2):93-7; Warren et al., 2000, Clin Lymphoma, 1(1):57-61, which are incorporated herein by reference).
  • Antibodies of the invention can be used in combination with a therapeutic antibody that does not mediate its therapeutic effect through cell killing to potentiate the antibody's therapeutic activity.
  • the invention encompasses use of the
  • NYJD 1603354.2 antibodies of the invention in combination with a therapeutic apoptosis inducing antibody with agonisitc activity, e.g., an anti-Fas antibody.
  • Anti-Fas antibodies are known in the art and include for example, Jo2 (Ogasawara et al, 1993, Nature 364: 806)and HFE7 (Ichikawa et al, 2000, Int. Immunol. 12: 555).
  • Jo2 (Ogasawara et al, 1993, Nature 364: 806)and HFE7 (Ichikawa et al, 2000, Int. Immunol. 12: 555).
  • Fc)RIIB has been implicated in promoting anti-Fas mediated apoptosis, see, e.g., Xu et al, 2003, Journal of Immunology, 171: 562-568.
  • the extracellular domain of Fc ⁇ RIIB may serve as a cross-linking agent for Fas receptors, leading to a functional complex and promoting Fas dependent apoptosis.
  • the antibodies of the invention block the interaction of anti-Fas antibodies and Fc ⁇ RIIB, leading to a reduction in Fas-mediated apoptotic activity.
  • Antibodies of the invention that result in a reduction in Fas-mediated apoptotic activity are particularly useful in combination with anti-Fas antibodies that have undesirable side effects, e.g., hepatotoxicity.
  • the antibodies of the invention enhance the interaction of anti-Fas antibodies and Fc)RIIB, leading to an enhancement of Fas-mediated apoptotic activity.
  • Combination of the antibodies of the invention with therapeutic apoptosis inducing antibodies with agonisitc activity have an enhanced therapeutic efficacy.
  • Therapeutic apoptosis inducing antibodies used in the methods of the invention may be specific for any death receptor known in the art for the modulation of apoptotic pathway, e.g., TNFR receptor family.
  • the invention provides a method of treating diseases with impaired apoptotic mediated signaling, e.g., cancer.
  • the invention encompasses a method of treating a disease with deficient Fas-mediated apoptosis, said method comprising administering an antibody of the invention in combination with an anti-Fas antibody.
  • the agonistic antibodies of the invention are particularly useful for the treatment of tumors of non-hematopoietic origin, including tumors of melanoma cells.
  • the efficacy of the agonistic antibodies of the invention is due, in part, to activation of Fc ⁇ RIIB inhibitory pathway, as tumors of non-hematopoietic origin, including tumors of melanoma cells express Fc ⁇ RIIB.
  • Fc ⁇ RIIB inhibitory pathway As tumors of non-hematopoietic origin, including tumors of melanoma cells express Fc ⁇ RIIB.
  • Recent experiments have in fact shown that expression of Fc ⁇ RIIB in melanoma cells modulates tumor growth by direct interaction with anti-tumor antibodies (e.g., by binding the Fc region of the anti-tumor antibodies) in an intracytoplasmic-dependent manner (Cassard et al., 2002, Journal of Clinical Investigation, 110(10): 1549-1557).
  • the invention encompasses use of the antibodies of the invention in combination with therapeutic antibodies that immunospecifically bind to tumor antigens that are not expressed on the tumor cells themselves, but rather on the surrounding reactive and tumor supporting, non-malignant cells comprising the tumor stroma.
  • the tumor stroma comprises endothelial cells forming new blood vessels and stromal fibroblasts surrounding the tumor vasculature.
  • an antibody of the invention is used in combination with an antibody that immunospecifically binds a tumor antigen on an endothelial cell.
  • an antibody of the invention is used in combination with an antibody that immunospecifically binds a tumor antigen on a fibroblast cell, e.g., fibroblast activation protein (FAP).
  • FAP fibroblast activation protein
  • FAP is a 95 KDa homodimeric type II glycoprotein which is highly expressed in stromal fibroblasts of many solid tumors, including, but not limited to lung, breast, and colorectal carcinomas.
  • IgE-targeted immediate hypersensitivity and allergic inflammation reactions have been proposed as possible natural mechanisms involved in anti-tumor responses (For a review see, e.g., Mills et al, 1992, Am. Journal of Epidemiol. 122: 66-74; Eriksson et al, 1995, Allergy 50: 718-722).
  • IgEs reduces tumor growth, leading in some instances to tumor rejection.
  • IgE loaded tumor cells not only possess a therapeutic potential but also confer long term antitumor immunity, including activation of innate immunity effector mechanism and T-cell mediated adaptive immune response, see Reali et al, 2001, Cancer Res.
  • the antagonistic antibodies of the invention may be used in the treatment and/or prevention of cancer in combination with administration of IgEs in order to enhance the efficacy of IgE-mediated cancer therapy. Although not intending to be bound by a particular mechanism of action the antibodies of the invention enhance the therapeutic efficacy of IgE treatment of tumors, by blocking the inhibitory pathway.
  • the antagonistic antibodies of the invention may enhance the therapeutic efficacy of IgE mediated cancer therapy by (i) enhancing the delay in tumor growth; (ii) enhancing the decrease in the rate of tumor
  • NYJD 1603354.2 progression; (iii) enhancing tumor rejection; or (iv) enhancing protective immune relative to treatment of cancer with IgE alone.
  • Cancer therapies and their dosages, routes of administration and recommended usage are known in the art and have been described in the literature, see, e.g., Physician's Desk Reference (56 th ed., 2002, which is incorporated herein by reference).
  • the agonistic antibodies of the invention are useful for treating or preventing any
  • B cell malignancies particularly non-Hodgkin's lymphoma and chronic lymphocytic leukemia.
  • Fc ⁇ RIIB is a target for deregulation by chromosomal translocation in malignant lymphoma, particularly in B-cell non-Hodgkin's lymphoma ⁇ See Callanan M.B. et at, 2000 Proc. Natl. Acad. ScL U.S.A., 97(l):309-314).
  • the antibodies of the invention are useful for treating or preventing any chronic lymphocytic leukemia of the B cell lineage.
  • Chronic lymphocytic leukemia of the B cell lineage are reviewed by Freedman (See review by Freedman, 1990, Hemtaol.
  • the agonistic antibodies of the invention inhibit or prevent B cell malignancies inhibiting B cell proliferation and/or activation.
  • the invention also encompasses the use of the agonistic antibodies of the invention in combination with other therapies known (e.g., chemotherapy and radiotherapy) in the art for the prevention and/or treatment of B cell malignancies.
  • the invention also encompasses the use of the agonistic antibodies of the invention in combination with other antibodies known in the art for the treatment and or prevention of B-cell malignancies.
  • the agonistic antibodies of the invention can be used in combination with the anti-C22 or anti-CD 19 antibodies disclosed by Goldenberg et al. (U.S. 6,306,393).
  • Antibodies of the invention can also be used in combination with for example but not by way of limitation, Oncoscint (target: CEA), Verluma (target: GP40), Prostascint (target: PSMA), CEA-SCAN(target: CEA), Rituxin (target: CD20), Herceptin (target: HER-2), Campath (target: CD52), Mylotarge (target: CD33), and Zevalin (target: CD20).
  • Oncoscint target: CEA
  • Verluma target: GP40
  • Prostascint target: PSMA
  • CEA-SCAN target: CEA
  • Rituxin target: CD20
  • Herceptin target: HER-2
  • Campath target: CD52
  • Mylotarge target: CD33
  • Zevalin target: CD20
  • the invention provides methods for treating or preventing an IgE-mediated and or Fc ⁇ RI mediated allergic disorder in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of the agonistic antibodies or fragments thereof of the invention.
  • antibodies of the invention are useful in inhibiting Fc ⁇ RI-induced mast cell activation, which contributes to acute and late phase allergic responses (Metcalfe D. et al. 1997, Physiol. Rev. 77:1033).
  • the agonistic antibodies of the invention have enhanced therapeutic efficacy and/or reduced side effects in comparison with the conventional methods used in the art for the treatment and/or prevention of IgE mediated allergic disorders.
  • Conventional methods for the treatment and/or prevention of IgE mediated allergic disorders include, but are not limited to, anti-inflammatory drugs (e.g., oral and inhaled corticosteroids for asthma), antihistamines (e.g., for allergic rhinitis and atopic dermatitis), cysteinyl leukotrienes (e.g., for the treatment of asthma); anti-IgE antibodies; and specific immunotherapy or desensitization.
  • anti-inflammatory drugs e.g., oral and inhaled corticosteroids for asthma
  • antihistamines e.g., for allergic rhinitis and atopic dermatitis
  • cysteinyl leukotrienes e.g., for the treatment of asthma
  • anti-IgE antibodies e.g., specific immunotherapy or desensitization.
  • IgE-mediated allergic responses include, but are not limited to, asthma, allergic rhinitis, gastrointestinal allergies, eosinophilia, conjunctivitis, atopic dermatitis, urticaria, anaphylaxis, or golmerular nephritis.
  • the invention encompasses molecules, e.g., immunoglobulins, engineered to form complexes with Fc ⁇ RI and human Fc ⁇ RIIB, i.e., specifically bind Fc ⁇ RI and human Fc)RIIB.
  • molecules e.g., immunoglobulins
  • Fc ⁇ RI and human Fc ⁇ RIIB i.e., specifically bind Fc ⁇ RI and human Fc)RIIB.
  • such molecules have therapeutic efficacy in IgE and Fc ⁇ RI-mediated disorders.
  • the therapeutic efficacy of these engineered molecules is, in part, due to their ability to inhibit mast cell and basophil function.
  • Fc ⁇ RIIB are chimeric fusion proteins comprising a binding site for Fc ⁇ RI and a binding site for Fc ⁇ RIIB. Such molecules may be engineered in accordance with standard recombinant DNA methodologies known to one skilled in the art.
  • a chimeric fusion protein for use in the methods of the invention comprises an F(ab') single chain of an anti-Fc ⁇ RIIB monoclonal antibody of the invention fused to a region used as a bridge to link the huFc ⁇ to the C-terminal region of the F(ab') single chain of the anti-Fc ⁇ RIIB monoclonal antibody.
  • One exemplary chimeric fusion protein for use in the methods of the invention comprises the following: V 1 ZC H (Fc ⁇ RIIB )- hinge- V H /C H (Fc ⁇ RIIB )-LINKER -C H ⁇ 2-C H ⁇ 3- C ⁇ 4.
  • the linker for the chimeric molecules may be five, ten, preferably fifteen amino acids in length. The length of the linker may vary to provide optimal binding of the molecule to both Fc ⁇ RIIB and Fc ⁇ RI. In a specific embodiment, the linker is a 15 amino acid linker, consisting of the sequence: (Gly 4 Ser) 3 .
  • the flexible peptide linker facilitates chain pairing and minimizes possible refolding and it will also allow the chimeric molecule to reach the two receptors, i.e., Fc ⁇ RIIB and Fc ⁇ RI on
  • NYJD 1603354 2 the cells and cross-link them.
  • the chimeric molecule is cloned into a mammalian expression vector, e.g., pCI-neo, with a compatible promoter, e.g., cytomegalovirus promoter.
  • the fusion protein prepared in accordance with the methods of the invention will contain the binding site for Fc ⁇ RI (CH ⁇ 2CH ⁇ 3) and for Fc ⁇ RIIB (VL/CL,- hinge- VH/CH).
  • the nucleic acid encoding the fusion protein prepared in accordance with the methods of the invention is preferably transfected into 293 cells and the secreted protein is purified using common methods known in the art.
  • Binding of the chimeric molecules to both human Fc ⁇ RI and Fc ⁇ RIIB may be assessed using common methods known to one skilled in the art for determining binding to an Fc)R.
  • the chimeric molecules of the invention have therapeutic efficacy in treating IgE mediated disorders, for example, by inhibiting antigen-driven degranulation and inhibition of cell activation.
  • the efficacy of the chimeric molecules of the invention in blocking IgE driven Fc ⁇ RI-mediated mast cell degranulation may be determined in transgenic mice, which have been engineered to express the human Fc ⁇ R ⁇ and human Fc ⁇ RIIB, prior to their use in humans.
  • the invention provides the use of bispecific antibodies for the treatment and/or prevention of IgE-mediated and/or Fc ⁇ RI-mediated allergic disorders.
  • a bispecific antibody (BsAb) binds to two different epitopes usually on distinct antigens. BsAbs have potential clinical utility and they have been used to target viruses, virally infected cells and bacterial pathogens as well as to deliver thrombolitic agents to blood clots (Cao Y., 1998 Bioconj. Chem 9: 635-644; Koelemij et al, 1999, /. Immunother., 22, 514-524; Segal et al, Curr. Opin. Immunol. , 11, 558-562).
  • the technology for the production of BsIgG and other related bispecific molecules is available (see, e.g., Carter et al., 2001 J. of Immunol. Methods, 248, 7-15; Segal et al, 2001, J. of Immunol. Methods, 248, 7-15, which are incorporated herein by reference in their entirety).
  • the instant invention provides bispecific antibodies containing one F(ab')of the anti- Fc ⁇ RIlB antibody and one F(ab') of an available monoclonal anti-hulgE antibody which aggregates two receptors, Fc ⁇ RIIB and Fc ⁇ RI, on the surface of the same cell. Any methodology known in the art and disclosed herein may be employed to generate bispecific antibodies for use in the methods of the invention.
  • the BsAbs will be produced by chemically cross-linking F(ab') fragments of an anti-Fc ⁇ RUB antibody and an anti- hulgE antibody as described previously, see, e.g., Glennie et al, 1995, Tumor Immunobiologv, Oxford University press, Oxford, p. 225; which is incorporated herein by reference in its entirety).
  • the F(ab') fragments may be produced by limited proteolysis with pepsin and reduced
  • NYJD 1603354.2 with mercaptoethanol amine to provide Fab' fragments with free hinge-region sulfhydryl (SH) groups.
  • the SH group on one of the Fab' (SH) fragments may be alkylated with excess 0- phenylenedimaleimide (0-PDM) to provide a free maleimide group (mal).
  • the two preparations Fab'(mal) and Fab'(SH) may be combined at an appropriate ratio, preferably 1:1 to generate heterodimeric constructs.
  • the BsAbs can be purified by size exclusion chromatography and characterized by HPLC using methods known to one skilled in thr art.
  • the invention encompasses bispecific antibodies comprising a first heavy chain-light chain pair that binds Fc ⁇ RIIB with greater affinity than said heavy chain-light chain pair binds Fc ⁇ RIIA, and a second heavy chain-light chain pair that binds IgE receptor, with the provision that said first heavy chain-light chain pair binds Fc ⁇ RIIB first.
  • the bispecific antibodies of the invention can be engineered using standard techniques known in the art to ensure that the binding to Fc ⁇ RIIB precedes the binding to the IgE receptor. It will be understood to one skilled in the art to engineer the bispecific antibodies, for example, such that said bispecific antibodies bind Fc ⁇ RIIB with greater affinity than said antibodies bind IgE receptor.
  • the bispecific antibodies can be engineered by techniques known in the art, such that the hinge size of the antibody can be increased in length, for example, by adding linkers, to provide the bispecific antibodies with flexibility to bind the IgE receptor and Fc ⁇ RIIB receptor on the same cell.
  • the antibodies of the invention can also be used in combination with other therapeutic antibodies or drugs known in the art for the treatment or prevention of IgE-mediated allergic disorders.
  • the antibodies of the invention can be used in combination with any of the following: azelastine, Astelin, beclomethasone dipropionate inhaler, Vanceril, beclomethasone dipropionate nasal inhaler/spray, Vancenase, Beconase budesonide nasal inhaler/spray, Rhinocort cetirizine, Zyrtec chlorpheniramine, pseudoephedrine, Deconamine, Sudafed, cromolyn, Nasalcrom, Intal, Opticrom, desloratadine, Clarinex, fexofenadine and pseudoephedrine, Allegra-D, fexofenadine, Allegra flunisolide nasal spray, Nasalide fluticasone propionate nasal inhaler/spray, Fl
  • Antibodies of the invention can be used in combination with cytosine-guanine dinucleotides (“CpG”)-based products that have been developed (Coley Pharmaceuticals) or are currently being developed as activators of innate and acquired immune responses.
  • CpG cytosine-guanine dinucleotides
  • the invention encompasses the use of CpG 7909,
  • NYJD 16033542 CpG 8916, CpG 8954 (Coley Pharmaceuticals) in the methods and compositions of the invention for the treatment and/or prevention of IgE-mediated allergic disorders (See also Weeratna et al, 2001, FEMS Immunol Med Microbiol, 32(1):65-71, which is incorporated herein by reference).
  • the invention encompasses the use of the antibodies of the invention in combination with any therapeutic antibodies known in the art for the treatment of allergy disorders, e.g., XolairTM (Omalizumab; Genentech); rhuMAB-E25 (Bio World Today, Nov. 10, 1998, p. 1; Genentech); CGP-51901 (humanized anti-IgE antibody), etc.
  • the invention encompasses the use of the antibodies of the invention in combination with other compositions known in the art for the treatment of allergy disorders. In particular methods and compositions disclosed in Carson et a (US 6,426,336; US 2002/0035109 Al; US 2002/0010343) is incorporated herein by reference in its entirety.
  • the methods of the invention encompass the administration of one or more angiogenesis inhibitors such as but not limited to: Angiostatin (plasminogen fragment); antiangiogenic antithrombin III; Angiozyme; ABT-627; Bay 12-9566; Benefin; Bevacizumab; BMS-275291; cartilage-derived inhibitor (CDI); CAI; CD59 complement fragment; CEP-7055; Col 3; Combretastatin A-4; Endostatin (collagen XVIII fragment); Fibronectin fragment; Gro-beta; Halofuginone; Heparinases; Heparin hexasaccharide fragment; HMV833; Human chorionic gonadotropin (hCG); IM-862; Interferon alpha/beta/gamma; Interferon inducible protein (IP-10); Interleukin-12; Kringle 5 (plasminogen fragment); Marimastat; Metalloproteinase inhibitors (TIMP
  • Anti-cancer agents that can be used in combination with antibodies of the invention in the various embodiments of the invention, including pharmaceutical compositions and dosage forms and kits of the invention, include, but are not limited to: acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin;
  • NYJD 1603354.2 sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride.
  • Other anti-cancer drugs include, but are not limited to: 20-epi-l,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein- 1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphi
  • NYJD: 1603354.2 is a primatized anti-CD4 antibody (IDEC); IDEC- 152 is a primatized anti-CD23 antibody (IDEC/Seikagaku); SMART anti-CD3 is a humanized anti-CD3 IgG (Protein Design Lab); 5Gl.1 is a humanized anti-complement factor 5 (C5) antibody (Alexion Pharm); D2E7 is a humanized anti-TNF- ⁇ antibody (CAT/BASF); CDP870 is a humanized anti-TNF- ⁇ Fab fragment (Celltech); IDEC- 151 is a primatized anti-CD4 IgGl antibody (IDEC Pharm/SmithKline Beecham); MDX-CD4 is a human anti-CD4 IgG antibody (Medarex/Eisai/Genmab); CDP571 is a humanized anti-TNF- ⁇ IgG4 antibody (Celltech); LDP- 02 is a humanized anti- ⁇ 4 ⁇ 7 antibody (Le
  • Table 4 Monoclonal antibodies for Cancer Therapy that can be used in combination with the antibodies of the invention.
  • Company Product Disease Target Monoclonal antibodies for Cancer Therapy that can be used in combination with the antibodies of the invention.
  • Herceptin metastatic breast HER-2 cancer Herceptin early stage HER-2 breast cancer
  • IDEC Zevalin (Rituxan + low grade of CD20 yttrium-90) follicular, relapsed or refractory, CD20-positive, B-cell NHL and Rituximab- refractory NHL
  • Cetuximab + cisplatin head & neck EGF receptor cancer extendensive incurable local- regional disease & distant metasteses
  • IMC-ICl 1 colorectal cancer VEGF-receptor with liver metasteses
  • CEA-Scan Tc-99m- colorectal cancer CEA labeled arcitumomab (radioimaging)
  • CEA-Scan Tc-99m- Breast cancer CEA labeled arcitumomab (radioimaging)
  • CEA-Scan Tc-99m- lung cancer CEA labeled arcitumomab (radioimaging)
  • CEA-Scan Tc-99m- intraoperative CEA labeled arcitumomab tumors (radio imaging)
  • LymphoScan Tc-99m- lymphomas CD22 labeled (radioimaging)
  • CTLA-4 Medarex MDX-101 (CTLA-4) Prostate and CTLA-4 other cancers
  • NeoRx CD20-streptavidin (+ Non-Hodgkins CD20 biotin- yttrium 90) lymphoma
  • MAb lung/kidney lung & kidney NA cancer cancer nacolomab tafenatox colon & NA
  • GlioMAb-H (+ gelonin glioma, NA toxin) melanoma & neuroblastoma
  • the invention provides a method for enhancing an immune response to a vaccine composition in a subject, said method comprising administering to said subject an antibody or a fragment thereof that specifically binds Fc ⁇ RIIB with greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA, and a vaccine composition, wherein said antibody or a fragment thereof enhances the immune response to said vaccine composition.
  • said antibody or a fragment thereof enhances the immune response to said vaccine composition by enhancing antigen presentation/and or antigen processing of the antigen to which the vaccine is directed at. Any vaccine composition known in the art is useful in combination with the antibodies or fragments thereof of the invention.
  • the invention encompasses the use of the antibodies of the invention in combination with any cancer vaccine known in the art, e.g., CanvaxinTM (Cancer Vax, Corporation, melanoma and colon cancer); Oncophage (HSPPC-96; Antigenics; metastatic melanoma); HER-2/neu cancer vaccine, etc.
  • the cancer vaccines used in the methods and compositions of the invention can be, for example, antigen-specific vaccines, anti-idiotypic vaccines, dendritic cell vaccines, or DNA vaccines.
  • the invention encompasses the use of the antibodies of the invention with cell-based vaccines as described by Segal et al. (US 6,403,080), which is incorporated herein by reference in its entirety.
  • the cell based vaccines used in combination with the antibodies of the invention can be either autologous or allogeneic.
  • the cancer-based vaccines as described by Segal et al. are based on Opsonokine (TM) product by Genitrix, LLC.
  • Opsonokines(TM) are genetically engineered cytokines that, when mixed with tumor cells, automatically attach to the surface of the cells.
  • the cytokine on the cells activates critical antigen presenting cells in the recipient, while also allowing the antigen presenting cells to ingest the tumor cells.
  • the antigen presenting cells are then able to instruct "killer" T cells to find and destroy similar tumor cells throughout the body.
  • the Opsonokine(TM) product converts the tumor cells into a potent anti-tumor immunotherapeutic.
  • the invention encompasses the use of the antibodies of the invention in combination with any allergy vaccine known in the art.
  • the antibodies of the invention can be used, for example, in combination with recombinant hybrid molecules coding for the major timothy grass pollen allergens used for vaccination against grass pollen allergy, as described by Linhart et al. (2000, FASEB Journal, 16(10): 1301-3, which is incorporated by reference).
  • the antibodies of the invention can be used in combination with DNA- based vaccinations described by Horner et al. (2002, Allergy, 57 Suppl, 72:24-9, which is incorporated by reference).
  • Antibodies of the invention can be used in combination with Bacille Clamett-Guerin (“BCG”) vaccination as described by Choi et al. (2002, Ann. Allergy Asthma Immunology, 88(6): 584-91) and Barlan et al. (2002, Journal Asthma, 39(3):239-46), both of which are incorporated herein by reference in entirety, to downregulate IgE secretion.
  • BCG Bacille Clamett-Guerin
  • the antibodies of the invention are useful in treating food allergies.
  • the antibodies of the invention can be used in combination with vaccines or other immunotherapies known in the art (see Hourihane et al., 2002, Curr. Opin. Allergy Clin. Immunol. 2(3):227-31) for the treatment of peanut allergies
  • the methods and compositions of the invention can be used in combination with vaccines, in which immunity for the antigen(s) is desired.
  • antigens may be any antigen known in the art.
  • the antibodies of the invention can be used to enhance an immune response, for example, to infectious agents, diseased or abnormal cells such as, but not limited to, bacteria (e.g., gram positive bacteria, gram negative bacteria, aerobic bacteria, Spirochetes, Mycobacteria, Rickettsias, Chlamydias, etc.), parasites, fungi (e.g., Candida albicans, Aspergillus, etc.), viruses (e.g., DNA viruses, RNA viruses, etc.), or tumors.
  • bacteria e.g., gram positive bacteria, gram negative bacteria, aerobic bacteria, Spirochetes, Mycobacteria, Rickettsias, Chlamydias, etc.
  • fungi e.g., Candida albicans, Asper
  • Viral infections include, but are not limited to, human immunodeficiency virus (HIV); hepatitis A virus, hepatitis B virus, hepatitis C virus, hepatitis D virus, or other hepatitis viruses; cytomagaloviruses, herpes simplex virus-1 (-2,-3,-4,-5,-6), human papilloma viruses; Respiratory syncytial virus (RSV), Parainfluenza virus (PIV), Epstein Barr virus, or any other viral infections.
  • the invention encompasses the use of the antibodies of the invention to enhance a humoral and/or cell mediated response against the antigen(s) of the vaccine composition.
  • the invention further encompasses the use of the antibodies of the invention to either prevent or treat a particular disorder, where an enhanced immune response against a particular antigen or antigens is effective to treat or prevent the disease or disorder.
  • diseases and disorders include, but are not limited to, viral infections, such as HIV, CMV, hepatitis, herpes virus, measles, etc., bacterial infections, fungal and parasitic infections, cancers, and any other disease
  • NYJD 1603354.2 or disorder amenable to treatment or prevention by enhancing an immune response against a particular antigen or antigens.
  • Certain cancers may be associated with an ability of the tumors to circumvent an immune response against their antigens, i.e., tolerance to these antigens exists. See Mapara et al., 2004, J. Clin. Oncol. 22:1136-1151. Accordingly, a goal in tumor immunotherapy is to break tolerance to tumor antigens in order to induce an antitumor response. Eliciting an immune response against a foreign antigen that is otherwise recognized by the host as a "self antigen breaks tolerance to that antigen.
  • the invention provides a method for breaking tolerance to an antigen in a patient by administering to a patient in need thereof (1) an antigen- antibody complex comprising the antigen and (2) an antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB, thereby breaking tolerance in said patient to the antigen.
  • the antibody or fragment thereof can be administered before, concurrently with, or after administration of said antigen-antibody complex.
  • Antigen-presenting cells such as dendritic cells, coexpress activating and inhibitory Fc gamma receptors.
  • Fc Fc
  • Antigen-presenting cells such as dendritic cells
  • the antigen-antibody complexes comprising an antigen are primarily taken up by non-inhibitory receptors on antigen-presenting cells elicting an immune response to the antigen.
  • the antigen is an antigen that is associated with a cancer or a neoplastic disease.
  • the antigen is specific to a cancer cell or a neoplastic cell.
  • the antigen can also be an antigen of a pathogen, such as, e.g., a virus, a bacterium, or a protozoa. Representative antigens have been disclosed herein.
  • the invention provides methods and pharmaceutical compositions comprising antibodies of the invention.
  • the invention also provides methods of treatment, prophylaxis, and amelioration of one or more symptoms associated with a disease, disorder or infection by administering to a subject an effective amount of a fusion protein or a conjugated molecule of the invention, or a pharmaceutical composition comprising a fusion protein or conjugated molecules of the invention.
  • NYJD 16033542 molecule is substantially purified (i.e., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is an animal, preferably a mammal such as non-primate (e.g., cows, pigs, horses, cats, dogs, rats etc.) and a primate (e.g., monkey such as, a cynomolgous monkey and a human).
  • the subject is a human.
  • compositions comprising antibodies of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or fusion protein, receptor- mediated endocytosis (See, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • antibodies of the invention e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or fusion protein, receptor- mediated endocytosis (See, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • the antibodies of the invention are formulated in liposomes for targeted delivery of the antibodies of the invention.
  • Liposomes are vesicles comprised of concentrically ordered phopsholipid bilayers which encapsulate an aqueous phase. Liposomes typically comprise various types of lipids, phospholipids, and/or surfactants. The components of liposomes are arranged in a bilayer configuration, similar to the lipid arrangement of biological membranes. Liposomes are particularly preferred delivery vehicles due, in part, to their biocompatibility, low immunogenicity, and low toxicity.
  • the invention also encompasses methods of preparing liposomes with a prolonged serum half-life, i.e., enhanced circulation time, such as those disclosed in U.S. Patent No. 5,013,556.
  • Preferred liposomes used in the methods of the invention are not rapidly cleared from circulation, i.e., are not taken up into the mononuclear phagocyte system (MPS).
  • MPS mononuclear phagocyte system
  • the invention encompasses sterically stabilized liposomes which are prepared using common methods known to one skilled in the art.
  • sterically stabilized liposomes contain lipid components with bulky and highly flexible hydrophilic moieties, which reduces the unwanted reaction of liposomes with serum proteins, reduces oposonization with serum components and reduces recognition by MPS.
  • Sterically stabilized liposomes are preferably prepared using polyethylene glycol.
  • For preparation of liposomes and sterically stabilized liposome see, e.g., Bendas et ah, 2001 BioDrugs, 15(4): 215-224; Allen et ai, 1987 FEBS Lett.
  • Liposome Res. 6 99-116; Litzinger et al, 1994, Biochim. Biophys. Acta, 1190: 99-107; Maruyama et al, 1991, Chem. Pharm. Bull., 39: 1620-2; Klibanov et al., 1991, Biochim Biophys Acta, 1062; 142-8; Allen et al, 1994, Adv. Drug Deliv. Rev, 13: 285-309; all of which are incorporated herein by reference in their entirety.
  • the invention also encompasses liposomes that are adapted for specific organ targeting, see, e.g., U.S. Patent No. 4,544,545.
  • Particularly useful liposomes for use in the compositions and methods of the invention can be generated by reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG derivatized phosphatidylethanolamine (PEG-PE).
  • PEG-PE PEG derivatized phosphatidylethanolamine
  • Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • a fragment of an antibody of the invention e.g., F(ab') may be conjugated to the liposomes using previously described methods, see, e.g., Martin et al., 1982, J. Biol. Chem. 257: 286-288, which is incorporated herein by reference in its entirety.
  • the antibodies of the invention may also be formulated as immunoliposomes.
  • Immunoliposomes refer to a liposomal composition, wherein an antibody of the invention or a fragment thereof is linked, covalently or non-covalently to the liposomal surface.
  • the chemistry of linking an antibody to the liposomal surface is known in the art and encompassed within the invention, see, e.g., Allen et al, 1995, Stealth Liposomes, Boca Rotan: CRC Press, 233-44; Hansen et al, 1995, Biochim. Biophys. Acta, 1239: 133-44; which are incorporated herein by reference in their entirety.
  • immunoliposomes for use in the methods and compositions of the invention are further sterically stabilized.
  • the antibodies of the invention are linked covalently or non-covalently to a hydrophobic anchor, which is stably rooted in the lipid bilayer of the liposome.
  • hydrophobic anchors include but are not limited to phospholipids, e.g., phosoatidylethanolamine (PE), phospahtidylinositol (PI).
  • PE phosoatidylethanolamine
  • PI phospahtidylinositol
  • any of the known biochemical strategies in the art may be used, see, e.g., J. Thomas August, ed., 1997, Gene Therapy: Advances in Pharmacology, Volume 40, Academic Press, San Diego, CA., p.
  • a functional group on an antibody molecule may react with an active group on a liposome associated hydrophobic anchor, e.g., an amino group of a lysine side chain on an antibody may be coupled to liposome associated N-glutaryl-phosphatidylethanolamine activated with water-soluble carbodiimide; or a thiol group of a reduced antibody can be coupled to liposomes via thiol reactive anchors such as pyridylthiopropionyl- phosphatidylethanolamine.
  • immunoliposomal formulations comprising an antibody of the invention are particularly effective as therapeutic agents, since they deliver the antibody to the cytoplasm of the target cell, i.e., the cell comprising the FcyRIIB receptor to which the antibody binds.
  • the immunoliposomes preferably have an increased half-life in blood, specifically target cells, and can be internalized into the cytoplasm of the target cells thereby avoiding loss of the therapeutic agent or degradation by the endolysosomal pathway.
  • the invention encompasses immunoliposomes comprising an antibody of the invention or a fragment thereof.
  • the immunoliposomes further comprise one or more additional therapeutic agents, such as those disclosed herein.
  • the immunoliposomal compositions of the invention comprise one or more vesicle forming lipids, an antibody of the invention or a fragment or derivative thereof, and optionally a hydrophilic polymer.
  • a vesicle forming lipid is preferably a lipid with two hydrocarbon chains, such as acyl chains and a polar head group.
  • vesicle forming lipids examples include phospholipids, e.g., phosphatidylcholine, phosphatidylethanolamine, phosphatidic acid, phosphatidylinositol, sphingomyelin, and glycolipids, e.g., cerebrosides, gangliosides. Additional lipids useful in the formulations of the invention are known to one skilled in the art and encompassed within the invention.
  • the immunoliposomal compositions further comprise a hydrophilic polymer, e.g., polyethylene glycol, and gnaglioside GMl, which increases the serum half life of the liposome.
  • Methods of administering an antibody of the invention include, but are not limited to, parenteral administration ⁇ e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous), epidural, and mucosal ⁇ e.g., intranasal and oral routes).
  • parenteral administration e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous
  • epidural e.g., intranasal and oral routes.
  • mucosal e.g., intranasal and oral routes.
  • the antibodies of the invention are administered intramuscularly,
  • NYJD 1603354 2 intravenously, or subcutaneously.
  • the compositions may be administered by any convenient route, for example, by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Patent Nos.
  • the invention also provides that the antibodies of the invention are packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of antibody.
  • the antibodies of the invention are supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted, e.g., with water or saline to the appropriate concentration for administration to a subject.
  • the antibodies of the invention are supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more preferably at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, or at least 75 mg.
  • the lyophilized antibodies of the invention should be stored at between 2 and 8°C in their original container and the antibodies should be administered within 12 hours, preferably within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted.
  • antibodies of the invention are supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the antibody, fusion protein, or conjugated molecule.
  • the liquid form of the antibodies are supplied in a hermetically sealed container at least 1 mg/ml, more preferably at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/kg, at least 25 mg/ml, at least 50 mg/ml, at least 100 mg/ml, at least 150 mg/ml, at least 200 mg/ml of the antibodies.
  • composition of the invention which will be effective in the treatment, prevention or amelioration of one or more symptoms associated with a disorder can be determined by standard clinical techniques.
  • dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the dosage administered to a patient is typically 0.0001 mg/kg to 100 mg/kg of the patient's body weight.
  • the dosage administered to a patient is between 0.0001 mg/kg and 20 mg/kg, 0.0001 mg/kg and 10 mg/kg, 0.0001 mg/kg and 5 mg/kg, 0.0001 and 2 mg/kg, 0.0001 and 1 mg/kg, 0.0001 mg/kg and 0.75 mg/kg, 0.0001 mg/kg and 0.5 mg/kg, 0.0001 mg/kg to 0.25 mg/kg, 0.0001 to 0.15 mg/kg, 0.0001 to 0.10 mg/kg, 0.001 to 0.5 mg/kg, 0.01 to 0.25 mg/kg or 0.01 to 0.10 mg/kg of the patient's body weight.
  • human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible. Further, the dosage and frequency of administration of antibodies of the invention or fragments thereof may be reduced by enhancing uptake and tissue penetration of the antibodies by modifications such as, for example, lipidation.
  • the dosage of the antibodies of the invention administered to a patient are 0.01 mg to 1000 mg/day, when used as single agent therapy.
  • the antibodies of the invention are used in combination with other therapeutic compositions and the dosage administered to a patient are lower than when said antibodies are used as a single agent therapy.
  • compositions of the invention may be administered locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • an implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • care must be taken to use materials to which the antibody or the fusion protein does not absorb.
  • compositions can be delivered in a vesicle, in particular a liposome (See Langer, Science 249:1527-1533 (1990); Treat et al, in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 3 17-327; see generally ibid.).
  • the compositions can be delivered in a controlled release or sustained release system. Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more antibodies of the invention. See, e.g., U.S.
  • a pump may be used in a controlled release system (See Langer, supra; Sefton, 1987, CRC CHt. Ref. Biomed. Eng. 14:20; Buchwald et al, 1980, Surgery 88:507; and Saudek et al, 1989, N. Engl. J. Med. 321:574).
  • polymeric materials can be used to achieve controlled release of antibodies (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, 1983, J., Macromol ScL Rev. Macromol. Chem. 23:61; See also Levy et al, 1985, Science 228:190; During et al, 1989, Ann. Neurol. 25:351; Howard et al, 1989, J. Neurosurg. 7 1:105); U.S. Patent No.
  • polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly( acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co- glycolides) (PLGA), and polyorthoesters.
  • a controlled release system can be placed in proximity of the therapeutic target (e.g., the lungs), thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • polymeric compositions useful as controlled release implants are used according to Dunn et al. (See U.S. 5,945,155). This particular method is based upon the therapeutic effect of the in situ controlled release of the bioactive material from the polymer system. The implantation can generally occur anywhere within the body of the patient in need of therapeutic treatment.
  • a non- polymeric sustained delivery system whereby a non-polymeric implant in the body of the subject is used as a drug delivery system.
  • the organic solvent of the implant Upon implantation in the body, the organic solvent of the implant will dissipate, disperse, or leach from the composition into surrounding tissue fluid, and the non-polymeric material will gradually coagulate or precipitate to form a solid, microporous matrix (See U.S. 5,888,533).
  • composition of the invention is a nucleic acid encoding an antibody
  • the nucleic acid can be administered in vivo to promote expression of its encoded antibody, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (See U.S. Patent No.
  • a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination.
  • the therapeutically or prophylactically effective dosage administered to a subject is typically 0.1 mg/kg to 200 mg/kg of the subject's body weight.
  • the dosage administered to a subject is between 0.1 mg/kg and 20 mg/kg of the subject's body weight and more preferably the dosage administered to a subject is between 1 mg/kg to 10 mg/kg of the subject's body weight.
  • the dosage and frequency of administration of antibodies of the invention may be reduced also by enhancing uptake and tissue penetration (e.g., into the lung) of the antibodies or fusion proteins by modifications such as, for example, lipidation.
  • Treatment of a subject with a therapeutically or prophylactically effective amount of antibodies of the invention can include a single treatment or, preferably, can include a series of treatments.
  • a subject is treated with antibodies of the invention in the range of between about 0.1 to 30 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks.
  • the pharmaceutical compositions of the invention are administered once a day, twice a day, or three times a day.
  • the pharmaceutical compositions are administered once a week, twice a
  • NYJD 1603354.2 week, once every two weeks, once a month, once every six weeks, once every two months, twice a year or once per year. It will also be appreciated that the effective dosage of the antibodies used for treatment may increase or decrease over the course of a particular treatment.
  • compositions of the invention include bulk drug compositions useful in the manufacture of pharmaceutical compositions (e.g., impure or non-sterile compositions) and pharmaceutical compositions (i.e., compositions that are suitable for administration to a subject or patient) which can be used in the preparation of unit dosage forms.
  • Such compositions comprise a prophylactically or therapeutically effective amount of a prophylactic and/or therapeutic agent disclosed herein or a combination of those agents and a pharmaceutically acceptable carrier.
  • compositions of the invention comprise a prophylactically or therapeutically effective amount of antibodies of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises of a therapeutically effective amount of an antibody or a fragment thereof that binds Fc ⁇ RIIB with a greater affinity than said antibody or a fragment thereof binds Fc ⁇ RIIA, a cytotoxic antibody that specifically binds a cancer antigen, and a pharmaceutically acceptable carrier.
  • said pharmaceutical composition further comprises one or more anti-cancer agents.
  • the pharmaceutical composition comprises (i) a therapeutically effective amount of an antibody or fragment thereof that specifically binds the extracellular domain of human Fc ⁇ RIIB and blocks the Fc binding site of human Fc ⁇ RIIB; (ii) a cytotoxic antibody that specifically binds a cancer antigen; and (iii) a pharmaceutically acceptable carrier.
  • the term "pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete), excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical
  • excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the compositions of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include, but are not limited to those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with captions such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • nucleic acids comprising sequences encoding antibodies or fusion proteins, are administered to treat, prevent or ameliorate one or more symptoms associated with a disease, disorder, or infection, by way of gene therapy.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded antibody or fusion protein that mediates a therapeutic or prophylactic effect.
  • Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described below.
  • a composition of the invention comprises nucleic acids ncoding an antibody, said nucleic acids being part of an expression vector that expresses the itibody in a suitable host.
  • nucleic acids have promoters, preferably gterologous promoters, operably linked to the antibody coding region, said promoter being tducible or constitutive, and, optionally, tissue-specific.
  • Licleic acid molecules are used in which the antibody coding sequences and any other desired jquences are flanked by regions that promote homologous recombination at a desired site in the ⁇ nome, thus providing for intrachromosomal expression of the antibody encoding nucleic acids Coller and Smithies, 1989, Proc. Natl. Acad. ScL USA 86:8932-8935; and Zijlstra et al., 1989, ature 342:435-438).
  • composition of the invention comprises nucleic
  • nucleic acids encoding a fusion protein
  • said nucleic acids being a part of an expression vector that ⁇ ression the fusion protein in a suitable host.
  • nucleic acids have promoters, eferabiy heterologous promoters, operably linked to the coding region of a fusion protein, said omoter being inducible or constitutive, and optionally, tissue-specific.
  • nucleic acid molecules are used in which the coding sequence of the fusion protein id any other desired sequences are flanked by regions that promote homologous recombination a desired site in the genome, thus providing for intrachromosomal expression of the fusion otein encoding nucleic acids.
  • nucleic acids into a subject may be either direct, in which case the bject is directly exposed to the nucleic acid or nucleic acid-carrying vectors, or indirect, in hich case, cells are first transformed with the nucleic acids in vitro, then transplanted into the bject. These two approaches are known, respectively, as in vivo or ex vivo gene therapy.
  • the nucleic acid sequences are directly administered in vo, where it is expressed to produce the encoded product.
  • microparticle bombardment ⁇ e.g., a gene gun; olistic, Dupont
  • coating lipids or cell-surface receptors or transfecting agents, capsulation in liposomes, microparticles, or microcapsules, or by administering them in ikage to a peptide which is known to enter the nucleus, by administering it in linkage to a
  • nucleic acid-ligand complexes can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (See, e.g., PCT Publications WO 92/06180; WO 92/22635; W092/20316; W093/14188; WO 93/20221).
  • the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. ScL USA 86:8932-8935; and Zijlstra et al, 1989, Nature 342:435-438).
  • viral vectors that contain nucleic acid sequences encoding an antibody or a fusion protein are used.
  • a retroviral vector can be used (See Miller et al, 1993, Meth. Enzymol. 217:581-599). These retroviral vectors contain the components necessary for the correct packaging of the viral genome and integration into the host cell DNA.
  • the nucleic acid sequences encoding the antibody or a fusion protein to be used in gene therapy are cloned into one or more vectors, which facilitates delivery of the nucleotide sequence into a subject.
  • retroviral vectors More detail about retroviral vectors can be found in Boesen et ah, (1994, Biotherapy 6:291-302), which describes the use of a retroviral vector to deliver the mdr 1 gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy.
  • Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al. , 1994, J. Clin. Invest. 93:644-651; Klein et al, 1994, Blood 83:1467-1473; Salmons and Gunzberg, 1993, Human Gene Therapy 4:129-141; and Grossman and Wilson, 1993, Curr. Opin. in Genetics and Devel. 3:110-114.
  • Adenoviruses are other viral vectors that can be used in gene therapy.
  • Adenoviruses are especially attractive vehicles for delivering genes to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson (Current Opinion in Genetics and Development 3:499-503, 1993, present a review of adenovirus-based gene therapy. Bout et al, (Human Gene Therapy, 5:3-10, 1994) demonstrated the use of adenovirus vectors to transfer genes to the respiratory epithelia of rhesus monkeys. Other instances of the use of adenoviruses in gene therapy can be found in Rosenfeld et al, 1991, Science 252:431-434; Rosenfeld et al, 1992, Cell 68:143-155;
  • Adeno-associated virus has also been proposed for use in gene therapy
  • Another approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection.
  • the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a subject.
  • the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any method known in the art, including but not limited to, transfection, electroporation, microinjection, infection with a viral or bacteriophage vector, containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcellmediated gene transfer, spheroplast fusion, etc.
  • Numerous techniques are known in the art for the introduction of foreign genes into cells ⁇ See, e.g., Loeffler and Behr, 1993, Meth. Enzymol. 217:599-618, Cohen et al, 1993, Meth. Enzymol.
  • the technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
  • the resulting recombinant cells can be delivered to a subject by various methods known in the art.
  • Recombinant blood cells ⁇ e.g., hematopoietic stem or progenitor cells
  • the amount of cells envisioned for use depends on the desired effect, patient state, etc., and can be determined by one skilled in the art.
  • Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood, fetal liver, etc.
  • the cell used for gene therapy is autologous to the subject.
  • nucleic acid sequences encoding an antibody or a fusion protein are introduced into the cells such that they are expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect.
  • stem or progenitor cells are used. Any stem and/or progenitor cells which can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention (See e.g., PCT Publication WO 94/08598; Stemple and Anderson, 1992, Cell 7 1:973-985; Rheinwald, 1980, Meth. Cell Bio.
  • the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding region, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • the invention provides a pharmaceutical pack or kit comprising one or more containers filled with antibodies of the invention. Additionally, one or more other prophylactic or therapeutic agents useful for the treatment of a disease can also be included in the pharmaceutical pack or kit.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • kits that can be used in the above methods.
  • a kit comprises one or more antibodies of the invention.
  • a kit further comprises one or more other prophylactic or therapeutic agents useful for the treatment of cancer, in one or more containers.
  • a kit further comprises one or more cytotoxic antibodies that bind one or more cancer antigens associated with cancer, hi certain embodiments, the other prophylactic or therapeutic agent is a chemotherapeutic.
  • the prophylactic or therapeutic agent is a biological or hormonal therapeutic.
  • compositions or prophylactic or therapeutic agents of the invention are preferably tested in vitro, e.g., in a cell culture system, and then in vivo, e.g., in an animal model organism, such as a rodent animal model system, for the desired therapeutic activity prior to use in humans.
  • assays which can be used to determine whether administration of a specific pharmaceutical composition is indicated, include cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise contacted with a pharmaceutical composition, and the effect of such composition upon the tissue sample is observed, e.g., inhibition of or decrease in growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation.
  • the tissue sample can be obtained by biopsy from the patient. This test allows the identification of the therapeutically most effective prophylactic or therapeutic molecule(s) for each individual patient. Alternatively, instead of culturing cells from a patient, therapeutic agents and methods may be screened using cells of a tumor or malignant cell line.
  • cell proliferation can be assayed by measuring 3 H-thymidine incorporation, by direct cell count, by detecting changes in transcriptional activity of known genes such as proto-oncogenes (e.g., fos, myc) or cell cycle markers; cell viability can be assessed by trypan blue staining, differentiation can be assessed visually based on changes in morphology, decreased growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation, etc.
  • proto-oncogenes e.g., fos, myc
  • cell cycle markers e.g., cell cycle markers
  • cell viability can be assessed by trypan blue staining
  • differentiation can be assessed visually based on changes in morphology, decreased growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation, etc.
  • Combinations of prophylactic and/or therapeutic agents can be tested in suitable animal model systems prior to use in humans.
  • animal model systems include, but are not limited to, rats, mice, chicken, cows, monkeys, pigs, dogs, rabbits, etc. Any animal system well- known in the art may be used.
  • combinations of prophylactic and/or therapeutic agents are tested in a mouse model system.
  • Such model systems are widely used and well-known to the skilled artisan.
  • Prophylactic and/or therapeutic agents can be administered repeatedly. Several aspects of the procedure may vary such as the temporal regime of administering the prophylactic and/or therapeutic agents, and whether such agents are administered separately or as an admixture.
  • Preferred animal models for use in the methods of the invention are for example, transgenic mice expressing Fc ⁇ R on mouse effector cells, e.g., any mouse model described in U.S. Patent No. 5,877,396 (which is incorporated herein by reference in its entirety).
  • mice for use in the methods of the invention include but are not limited to mice carrying human Fc ⁇ RIIIA, mice carrying human Fc ⁇ RIIA, mice carrying human Fc ⁇ RIIB and human Fc ⁇ RIIIA, mice carrying human Fc ⁇ RIIB and human Fc ⁇ RIIA. .
  • Toxicity and efficacy of the prophylactic and/or therapeutic protocols of the instant invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 5O (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 5 o/ED 5 o.
  • Prophylactic and/or therapeutic agents that exhibit large therapeutic indices are preferred. While prophylactic and/or therapeutic agents that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of the prophylactic and/or therapeutic agents for use in humans.
  • the dosage of such agents lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • the anti-cancer activity of the therapies used in accordance with the present invention also can be determined by using various experimental animal models for the study of cancer such as the SCID mouse model or transgenic mice or nude mice with human xenografts, animal models, such as hamsters, rabbits, etc. known in the art and described in Relevance of
  • compositions of the invention are preferably tested in vitro, and then in vivo, for the desired therapeutic or prophylactic activity, prior to use in humans.
  • Therapeutic agents and methods may be screened using cells of a tumor or malignant cell line.
  • cell proliferation can be assayed by measuring 3 H-thymidine incorporation, by direct cell count, by detecting changes in transcriptional activity of known genes such as proto-oncogenes (e.g., fos, myc) or cell cycle markers; cell viability can be assessed by trypan blue staining, differentiation can be assessed visually based on changes in morphology, decreased growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation, etc.
  • proto-oncogenes e.g., fos, myc
  • cell cycle markers e.g., cell cycle markers
  • cell viability can be assessed by trypan blue staining
  • differentiation can be assessed visually based on changes in morphology, decreased growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation, etc.
  • Compounds for use in therapy can be tested in suitable animal model systems prior to testing in humans, including but not limited to in rats, mice, chicken, cows, monkeys, rabbits, hamsters, etc., for example, the animal models described above. The compounds can then be used in the appropriate clinical trials.
  • any assays known to those skilled in the art can be used to evaluate the prophylactic and/or therapeutic utility of the combinatorial therapies disclosed herein for treatment or prevention of cancer.
  • Labeled antibodies of the invention can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders or infections.
  • the invention provides for the detection or diagnosis of a disease, disorder or infection comprising: (a) assaying the expression of Fc ⁇ RIIB in cells or a tissue sample of a subject using one or more antibodies that immunospecifically bind to Fc ⁇ RIIB; and (b) comparing the level of the antigen with a control level, e.g., levels in normal tissue samples, whereby an increase in the assayed level of antigen compared to the control level of the antigen is indicative of the disease, disorder or infection.
  • a control level e.g., levels in normal tissue samples
  • Antibodies of the invention can be used to assay Fc ⁇ RIIB levels in a biological sample using classical immunohistological methods as described herein or as known to those of skill in the art (e.g., see Jalkanen et al, 1985, J. Cell. Biol. 101:976-985; Jalkanen et al, 1987, J. Cell . Biol. 105:3087-3096). Other antibody-based methods useful for detecting protein gene
  • NYJD: 1603354.2 expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • Suitable antibody assay labels are known in the art and include enzyme labels, such as, alkaline phosphatase, glucose oxidase; radioisotopes, such as iodine ( 125 1, 131 D, carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 121 In), and technetium ( 99 Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine.
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • Suitable antibody assay labels include enzyme labels, such as, alkaline phosphatase, glucose oxidase; radioisotopes, such as iodine ( 125 1, 131 D, carbon ( 14 C), sulfur ( 35 S), trit
  • diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled antibody that immunospecifically binds to Fc ⁇ RIIB; b) waiting for a time interval following the administration for permitting the labeled antibody to preferentially concentrate at sites in the subject where Fc ⁇ RIIB is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled antibody in the subject, such that detection of labeled antibody above the background level indicates that the subject has the disease, disorder, or infection.
  • the antibody is labeled with an imaging moiety which is detectable using an imaging system known to one of skill in the art.
  • Background level can be determined by various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system.
  • the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images.
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99m Tc.
  • the labeled antibody will then preferentially accumulate at the location of cells which contain the specific protein.
  • In vivo tumor imaging is described in S. W. Burchiel et ai, "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments.” (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B. A.
  • the time interval following the administration for permitting the labeled molecule to preferentially concentrate at sites in the subject and for unbound labeled molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours or 6 to 12 hours. In another embodiment the time interval following administration is 5 to 20 days or 5 to 10 days.
  • monitoring of a disease, disorder or infection is carried out by repeating the method for diagnosing the disease, disorder or infection, for example, one
  • NYJD 1603354 2 month after initial diagnosis, six months after initial diagnosis, one year after initial diagnosis, etc.
  • Presence of the labeled molecule can be detected in the subject using methods known in the art for in vivo scanning. These methods depend upon the type of label used. Skilled artisans will be able to determine the appropriate method for detecting a particular label. Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
  • CT computed tomography
  • PET position emission tomography
  • MRI magnetic resonance imaging
  • sonography sonography
  • the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurston et al., U.S. Patent No. 5,441,050).
  • the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument. In another embodiment, the molecule is labeled with a positron emitting metal and is detected in the patient using positron emission-tomography. In yet another embodiment, the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI).
  • MRI magnetic resonance imaging
  • a mouse monoclonal antibody was produced from clones 3H7 or 2B6 with
  • mice generated in Dr. Ravetch Laboratory, Rockefeller University) were immunized with Fc ⁇ RIIB purified from supernatant of 293 cells that had been transfected with cDNA encoding the extracellular domain of the human Fc ⁇ RIIB receptor, residues 1-180. Hybridoma cell lines from spleen cells of these mice were produced and screened for antibodies that specifically bind Fc ⁇ RIIB with greater affinity than the antibodies bind Fc ⁇ RIIA.
  • Fc ⁇ RIIA or Fc ⁇ RIIB using ELISA assays.
  • the plate is coated with 100 ng/well of
  • NYJD 1603354.2 Fc ⁇ RIIA or Fc ⁇ RIIB.
  • the binding of the antibody to the specific receptor is detected with goat anti-mouse HRP conjugated antibody by monitoring the absorbance at 650 run.
  • the ability of the antibody from the hybridoma supernatant to block binding of aggregated IgG to Fc ⁇ RIIB is monitored.
  • the plate is blocked with the appropriate "blocking agent”, washed three times (200 ⁇ l/well) with wash buffer (PBS plus 0.1% Tween).
  • the plate is pre- incubated with hybridoma supernatant for 1 hour at 37 °C.
  • RBL-2H3 cells are transfected with human Fc ⁇ RIIB; cells are stimulated with various concentration of goat anti- mouse F(ab) 2 fragment ranging from 0.03 ⁇ g/mL to 30 ⁇ g/mL; sensitized with either mouse IgE alone (at 0.01 ⁇ g/mL) or with an anti- Fc ⁇ RIIB antibody. After 1 hour incubation at 37° temperature, the cells are spun down; the supernatant is collected; and the cells are lysed. The ⁇ -hexoaminidase activity released in the supernatant is determined in a colorometric assay using p-nitrophenyl N-acetyl- ⁇ D-glucoasminide. The release ⁇ -hexoaminidase activity is expressed as a percentage of the released activity relative to the total activity.
  • FACS ANALYSIS CHO cells, expressing Fc ⁇ RIIB are stained with various antibodies and analyzed by FACS. In one series of experiment, the cells are directly labeled to determine if the monoclonal antibodies recognize the receptor.
  • Cells are washed and the secondary antibodies are added, goat anti-mouse-FITC to detect the bound antibody and Streptavidin-PE conjugated to detect the bound aggregated biotinylated human IgG and incubated on ice for 30 minutes. Cells are washed and analyzed by FACS.
  • B Lymphocytes are stained to detect the presence of Fc ⁇ RIIB and CD20.
  • 200 ⁇ l of "buffy coat" for each sample is incubated on ice with 2 ⁇ g of isotype control or the monoclonal antibodies, 2B6 or 3H7.
  • Cells are washed once with PBS+1%BSA and incubated with l ⁇ l of goat anti mouse-PE antibody for 30 minutes on ice.
  • Cells are washed once and CD20-FITC antibody (2 ⁇ g) is added to the samples and incubated on ice for 30 minutes.
  • AU samples are washed with PBS+1%BSA once and the cells are analyzed by FACS.
  • SKBR-3 cells are labeled with bis(acetoxymethyl) 2,2':6',2"-terpyridine-t-6"-dicarboxylate (DELFIA BATDA Reagent, Perkin Elmer/Wallac).
  • BATDA reagent is added to the cells and the mixture is incubated at 37°C preferably under 5% CO 2 , for at least 30 minutes.
  • the cells are then washed with a physiological buffer, e.g., PBS with 0.125 mM sulfinpyrazole, and media containing 0.125 mM sulfinpyrazole.
  • the labeled target cells are added to effector cells, e.g., PBMC, to produce effecto ⁇ target ratios of approximately 50:1, 75:1, or 100:1.
  • PBMC is isolated by layering whole blood onto Ficoll-Hypaque (Sigma) and spinning at room temperature for 30 mins at 500 g.
  • the leukocyte layer is harvested as effectors for Europium- based ADCC assays.
  • Frozen or freshly isolated elutriated monocytes (Advanced Biotechnologies, MD) is used as effectors with the tumor target cell lines at varying effector to target ratio of 100:1 to 10:1 and the concentration of the antibodies is titrated from l-15 ⁇ g/ml.
  • Monocytes obtained as frozen stocks stimulated with cytokines is used as effector cells in ADCC assays. If frozen monocytes perform optimally they will be routinely used otherwise fresh cells will be used.
  • MDM will be prepared by treatment with cytokines GM-CSF or M- CSF that are known to enhance the viability and differentiation of monocytes in culture. MDM will be stimulated with cytokines and the expression of the various Fc ⁇ Rs (I, HA, HB, and IIIA) determined by FACS analysis.
  • the effector and target cells are incubated for at least two hours, and up to 16 hours, at 37°C, under 5% CO 2 in the presence of an anti-tumor antibody, specific for an antigen expressed on the target cells, Her2/neu, and in the presence or absence of an anti-Fc ⁇ RIIB antibody.
  • an anti-tumor antibody specific for an antigen expressed on the target cells, Her2/neu, and in the presence or absence of an anti-Fc ⁇ RIIB antibody.
  • a chimeric 4D5 antibody that has been engineered to contain the N297A mutation which is used as a negative control since this antibody binds the tumor target cells via its
  • NYJD 1603354.2 variable region. Loss of glycosylation at this site abolishes binding of the Fc region of the antibody to Fc ⁇ R.
  • Commercially available human IgG 1/k serves as an isotype control for the anti-Fc ⁇ RIIB antibody.
  • Cell supernatants are harvested and added to an acidic europium solution (e.g. , DELFIA Europium Solution, Perkin Elmer/Wallac).
  • the fluorescence of the Europium-TDA chelates formed is quantitated in a time-resolved fluorometer (e.g., Victor 1420, Perkin Elmer/Wallac).
  • MR Maximal release
  • SR spontaneous release
  • AICC antibody independent cellular cytotoxicity
  • Fc ⁇ RIIB The binding of crude 3H7 supernatant and purified 3H7 supernatant was measured (FIG. IB). In each case, the supernatant was supplied at a concentration of 70 ⁇ g/ml and diluted up to 6-fold. As shown in FIG. IB, upon saturating conditions, the 3H7 supernatant binds Fc ⁇ RIIB four times better than it binds Fc ⁇ RIIA. Upon purification with an protein G column, the absolute binding of the 3H7 supernatant to each immunogen improves. [00364] Blocking of aggregated human IgG binding to Fc ⁇ RIIB by the antibody produced from the 3H7 clone.
  • the antibody present in the hybridoma supernatant binds Fc ⁇ RIIB at the IgG binding site and blocks IgG binding, then the aggregated IgG cannot bind the receptor and hence no absorbance at 650 can be detected.
  • the antibody in effect is a "blocking agent" that blocks the IgG binding site on Fc ⁇ RIIB.
  • the ELISA was carried out with no blocking, with a control supernatant, and with supernatant from the 3H7
  • NYJD 16033542 clone As shown in FIG. 2, the 3H7 supernatant completely blocks IgG binding, since aggregated IgG cannot bind the receptor as evident from the lack of absorbance at 650 run. The control supernatant however fails to block IgG binding; aggregated IgG binds the receptor as evident by the reading at 650nm. The control supernatant behaves similarly to the condition where no blocking was done.
  • FcyRIIB, and Fc ⁇ RIIIA The direct binding of supernatant from the hybridoma cultures from the 3H7 cell line to Fc ⁇ RIIA, Fc ⁇ RIIIA and Fc ⁇ RIIB were compared using an ELISA assay (FIG. 4) .
  • the antibody produced from clone 3H7 has no affinity for Fc ⁇ RIIIA, and binds
  • Fc ⁇ RIIB with about 4 times greater affinity than it binds Fc ⁇ RIIA.
  • the antibody produced from clone 2B6 has minimal affinity for Fc ⁇ RIIA, whereas the other three commercially available antibodies bind Fc ⁇ RIIA in a saturatable manner and twice as much as the antibody from clone 2B6 binds Fc ⁇ RIIA (FIG. 5B).
  • Blocking of aggregated human IgG to Fc ⁇ RIIB by the antibody produced from clone 2B6 The ability of the antibody produced from clone 2B6 to block binding of the aggregated IgG to Fc ⁇ RIIB was investigated by a blocking ELISA assay and compared to that of the antibody produced by clone 3H7. As shown in FIG. 6A, the control supernatant does not bind Fc ⁇ RIIB on the IgG binding site and the aggregated IgG can bind the receptor and hence
  • FIG. 6B represents the data in a bar diagram.
  • the antibody produced from clone 2B6 effectively blocks the binding of aggregated IgG to the Fc ⁇ RIIB receptor in CHO cells since no staining is observed for biotinylated aggregated IgG after the cells were pre-incubated with the monoclonal antibody.
  • the cells are only stained in the lower right panel, indicating that most of the cells were bound to the monoclonal antibody from the 2B6 clone.
  • IgGl as the isotype control
  • Lymphocytes A double staining FACS assay was used to characterize the antibody produced from clones 2B6 and 3H7 in human B lymphocytes. Cells were stained with anti-CD20 antibody which was FITC conjugated, to select the B-lymphocyte population, as well as the antibodies produced from clone 3H7 and 2B6, labeled with goat anti-mouse peroxidase. The horizontal axis represents the intensity of the anti-CD20 antibody fluorescence and the vertical axis represents the intensity of the monoclonal antibody fluorescence. As shown in FIGS.
  • FIG. 8A shows the staining of the isotype control, mouse IgGl.
  • Fc ⁇ RIIB were stained with IgGl isotype control (FIG. 9A; left panel) or with supernatant from the 3H7 hybridoma (FIG. 9B; right panel).
  • Goat anti-mouse peroxidase conjugated antibody was used as a secondary antibody.
  • the cells were then analyzed by FACS; cells that are stained
  • NYJD: 1603354.2 with the supernatant from the 3H7 hybridoma show a strong fluorescence signal and a peak shift to the right; indicating the detection of Fc ⁇ RIIB in the CHO cells by the supernatant produced from the 3H7 hybridoma.
  • Cells stained with the supernatant from the 2B6 hybridoma also show a significant fluorescence, as compared to cells stained with IgGl, and a peak shift to the right, indicating the detection of Fc ⁇ RIIB in the CHO cells by the supernatant produced from the 2B6 hybridoma.
  • CHO cells expressing hyFc ⁇ RIIB were incubated with the anti CD32B antibodies, 2B6 or 3H7. Cells were washed and 9 ⁇ g/ml of aggregated human IgG were added to the cells on ice. The human aggregated IgG were detected with goat anti human-IgG GITC conjugated. Samples were analyzed by FACS cells labeled with 2B6 or 3H7 showed a significant fluorescence peak in the presence of aggregated human IgG (FIG. 10). 2BG antibody completely blocks binding of aggregated IgG as evidenced by the fluorescent peak shift to the left. Whereas the 3H7 antibody partially blocks binding of aggregated IgG as shown by the intermediate fluorescent peak.
  • the other antibodies, 1D5, 1F2, 2El, 2H9, and 2Dl 1 do not block binding of aggregated IgG.
  • the amount of each antibody bound to the receptor on the cells was also detected (inset) on a separate set of samples using a goat anti-mouse PE conjugated antibody.
  • FACS profiles using 2B6, 3H7, and IV.3 antibodies on human peripheral blood leukocyte The FACS profile of the anti-Fc ⁇ RIIB antibodies and IV.3 antibody shows their ability to discriminate between the two Fc ⁇ RII isoforms, HB and ILA expressed on the human hematopoietic cells.
  • IV.3 one of the first antibodies (commercially available) used to define Fc ⁇ RII, shows preferential binding to Fc ⁇ RIIA.
  • huPBL were stained with the anti-Fc ⁇ RIIB antibodies 2B6 and 3H7 and with IV.3, which preferentially (but not exclusively) recognizes the Fc ⁇ RIIA isoform of the receptor
  • leukocytes populations were selected based on FSC vs. SSC gating (FIG. 11) and identified with specific markets: CD20 (B cells), CD56 or CD16 (NK cells, lymphocyte gate), CD14 (monocytes) and CD16 (granulocytes, granulocyte gate) (FIG. 11).
  • CD20-positive cells (B cells) were uniformly stained with 2B6,
  • IV.3 also stained the majority of CD20-positive cells. No staining was observed for CD16/CD56-positive NK cells, while only a fraction of CD14-(monocytes) and CD16- (granulocytes) positive cells were stained with 2B6, 3H7. In contrast, IV.3 strongly stained the vast majority of CD-14-positive monocytes and the totality of CD16-positive granulocytes (FIG. 11).
  • Transfected RBL cells expressing Fc ⁇ RIIB were suspended in fresh media containing 0.01 ⁇ g/ml of murine anti-DNP IgE and plated in 96 well plates at a concentration of 2xlO 4 cells/well. After over-night incubation at 37°C in the presence of CO 2 , cells were washed twice with pre-warmed release buffer (10 mM HEPES, 137 mM NaCl, 2.7 mM KCl, 0.4 mM sodium phosphate monobasic, 5.6 mM glucose, 1.8 mM calcium chloride, 1.3 mM magnesium sulfate and 0.04% BSA, pH 7.4) and treated at 37°C with serial dilutions of BSA-DNP-FITC complexed with chimeric 4-4-20 antibody or BSA-DNP-FITC complexed with chimeric D265A 4-4-20 antibody in 100 ⁇ l buffer/well in the presence of 2B6 antibody, 1F2 antibody or murine IgGl
  • NYJD 1603354.2 activating receptors or combinations of inhibitory and activating receptors as described above.
  • the F(ab') 2 fragments of polyclonal goat anti-mouse IgG recognized the the light chain of the murine IgE bound to Fc ⁇ RI, aggregated these activating receptors, and ⁇ -hexosaminidase release, a marker for degranulation (Aketani et ai, 2001, Immunol. Lett. 75:185-9), increased with increasing IgE (FIG. 12B).
  • the F(ab') 2 fragment in effect, co- cross-linked the rat Fc ⁇ RI with CD32B and resulted in a significant decrease in ⁇ - hexosaminidase release when compared to sensitized cells preincubated with an irrelevant murine IgG 1 isotype control matched antibody.
  • the human inhibitory receptor, CD32B can induce a negative signal in rat basophilic cells, validating these transfectants as a model for the study of anti-human CD32B antibodies.
  • HuCD32B + RBL-2H3 cells were sensitized with a murine IgE anti-DNP monoclonal antibody.
  • the challenge antigen, BSA-DNP was further conjugated to FITC to provide additional epitopes recognized by a chimeric version of 4- 4-20, a murine anti-fluorescein antibody whose Fc portion had been substituted with human IgGi Fc to allow for optimal binding to human CD32B.
  • a chimeric version of 4-4-20 with a human IgGi Fc bearing a mutation in position 265 (asparagine to alanine) was also generated. This chimeric D265A 4-4-20 antibody lacks the ability to bind Fc ⁇ R's, including CD32B.
  • BSA- DNP-FITC induced a dose-dependent release of ⁇ -hexosaminidase from IgE-sensitized RBL- 2H3 cells (FIG. 13C).
  • BSA-DNP-FITC complexed with chimeric D265A 4-4-20 showing that BSA-DNP-FITC- chimeric D265A 4-4-20, as expected, was unable to recruit CD32B to the activating receptor.
  • BSA-DNP-FITC complexed with chimeric 4-4-20 a substantial reduction in ⁇ - hexosaminidase release was observed (FIG. 13B).
  • the polyvalent antigen is capable of
  • NYJD 1603354.2 aggregating Fc ⁇ RI with ensuing degranulation, while the surrogate antigen complexed with IgG co-aggregates CD32B resulting in diminished degranulation.
  • F(ab) 2 fragments of 2B6 where prepared and cells pre-incubated with 2B6 F(ab) 2 , prior to activation with the immunocomplexed antigen.
  • the percentage of ⁇ -hexosaminidase release was restored to the maximum levels observed in cells treated with the polyvalent antigen alone (FIG. 13C).
  • IGROV-I, OVCAR-8, and SKBR-3 cells express the Her2/neu antigen
  • cells were stained with either purified 4D5 or ch4D5 antibody on ice; the unbound antibody was washed out with PBS/BSA buffer containing sodium azide, and the binding of 4D5 or ch4D5 was detected by goat anti-mouse or goat anti-human antibody conjugated to PE (Jackson Laboratories), respectively.
  • An irrelevant IgGl antibody (Becton Dickinson) served as a control for non-specific binding.
  • the ovarian tumor cell lines express less Her2/neu antigens than the breast carcinoma cell line and evaluating these cell lines in parallel will determine the stringency of tumor clearance by an anti-Fc ⁇ RIIB antibody of the invention.
  • Human monocytes are the effector population involved in ADCC that express both activating and inhibitory receptors.
  • the expression of Fc ⁇ Rs was tested by FACS analysis using several lots of frozen monocytes as these cells will be adoptively transferred as effectors to investigate the role of ch2B6 in tumor clearance.
  • Commercially obtained frozen elutriated monocytes were thawed in basal medium containing 10% human AB serum and in basal medium with human serum and 25 - 50 ng/ml GM-CSF.
  • NYJD 16033542 (Pharaiingen), along with relevant isotype controls.
  • Ch4D5 mediates effective ADCC with ovarian and breast cancer cells lines using PBMC.
  • the ADCC activity of anti-Her2/neu antibody was tested in a europium based assay.
  • the ovarian cell line, IGROV-I, and the breast cancer cell line, SKBR-3, were used as labeled targets in a 4 hour assay with human PBL as effector cells.
  • FIGS. 16A and B indicate that ch4D5 is functionally active in mediating lysis of targets expressing Her2/neu.
  • the effect of an antibody of the invention on the ADCC activity of the anti-Her2/neu antibody is subsequently measured.
  • ADCC antibody dependent cell-mediated cytotoxicity
  • target cells from the CD32B expressing B-cell lymphoma lines, Daudi and Raji were labeled with the europium chelate bis(acetoxymethyl) 2,2':6',2"-terpyridine-6,6"-dicarboxylate (DELFIA BATDA Reagent, Perkin Elmer/Wallac).
  • DELFIA BATDA Reagent Perkin Elmer/Wallac.
  • the labeled target cells were then opsonized (coated) with either chimeric anti-CD32B (ch2B6) or aglycosylated chimeric anti- CD32B (ch2B6Agly) antibodies at the indicated concentrations as shown in FIGS. 18 and 19.
  • PBMC Peripheral blood mononuclear cells isolated by Ficoll-Paque (Amersham Pharmacia) gradient centrifugation, were used as effector cells (Effector to Target ratio of 75 tol). Following a 3.5 hour incubation at 37 0 C, 5%CO 2 , cell supernatants were harvested and added to
  • NYJD 1603354.2 an acidic europium solution (DELFIA Europium Solution, Perkin Elmer/Wallac).
  • DELFIA Europium Solution Perkin Elmer/Wallac
  • the fluorescence of the Europium-TDA chelates formed was quantitated in a time-resolved fluorometer (Victor 2 1420, Perkin Elmer/Wallac).
  • Maximal release (MR) and spontaneous release (SR) were determined by incubation of target cells with 2% Triton X-100 and media alone, respectively.
  • Antibody independent cellular cytotoxicity (AICC) was measured by incubation of target and effector cells in the absence of antibody. Each assay is performed in triplicate. The mean percentage specific lysis is calculated as: (ADCC - AICC)/(MR-SR) x 100.
  • ADCC in vitro against CD32B-expressing, B-cell lymphoma lines, Daudi and Raji, at concentrations greater than approximately 10 ng/ml. This activity is likely to be Fc-dependent since the aglycoslyated version of this antibody, ch2B6Agly, which is unable to interact with the Fc-receptors has reduced activity in this assay.
  • mice are maintained at BIOCON, Inc. Rockville, Maryland (see attached protocol). Mice are housed in Biosafety Level-2 facilities for the xenograft model using the ascites-derived ovarian cells and pleural effusion-derived breast cancer cells as sources of tumors. Mice are placed in groups of 4 for these experiments and monitored three times weekly. The weight of the mice and survival time are recorded and criteria for growing tumors is abdominal distention and palpable tumors. Mice showing signs of visible discomfort or that reach 5 grams in tumor weight are euthanized with carbon dioxide and autopsied. The antibody-treated animals are placed under observation for an additional two months after the control group.
  • IGROV-I cells subcutaneously gives rise to fast growing tumors while the intraperitoneal route induces a peritoneal carcinomatosis which kills the mice in 2 months. Since the IGROV-I cells form tumors within 5 weeks, at day 1 after tumor cell injection, monocytes as effectors are co-injected i.p. along with therapeutic antibodies ch4D5 and ch2B6 at 4 ⁇ g each per gm of mouse body weight (mbw) (Table 5). The initial injection is followed by weekly injections of antibodies for 4-6 weeks thereafter. Human effectors cells are replenished once in two weeks. A group of mice will receive no therapeutic antibody but will be injected with ch4D5 N297A and human IgGl as isotype control antibodies for the anti-tumor and ch2B6 antibody, respectively.
  • mice each are required for testing the role of an anti-Fc ⁇ RIIB antibody in tumor clearance with one target and effector combination, with two different combinations of the antibody concentrations.
  • These groups are A) tumor cells, B) tumor cells and monocytes, C) tumor cells, monocytes, anti-tumor antibody, ch4D5, D) tumor cells, monocytes, anti-tumor antibody ch4D5, and an anti-Fc ⁇ RIIB antibody, e.g., ch2B6, E) tumor cells, monocytes, and an anti-Fc ⁇ RIIB antibody, e.g., ch2B6, and F) tumor cells, monocytes, ch4D5 N297A, and human IgGl.
  • Various combination of antibody concentration can be tested in similar schemes.
  • the endpoint of the xenograft tumor model is determined based on the size of the tumors (weight of mice), survival time, and histology report for each group in Table 6. Mice are monitored three times a week; criteria for growing tumors are abdominal distention and presence of palpable masses in the peritoneal cavity. Estimates of tumor weight versus days after inoculation is calculated. Based on these three criteria from group D mice in Table 6 versus the other groups of mice will define the role of anti-FcyRIIB antibodies in enhancement of tumor clearance. Mice that show signs of visible pain or reach 5 grams of tumor weight are euthanized with carbon dioxide and autopsied. The antibody-treated animals are followed for two months after this time-point.
  • NYJD 1603354.2 the neoplastic cells. FACS analysis is used to determine the percentage of Her2/neu+ neoplastic cells in the clinical samples. Samples with high percentage of Her2/neu+ neoplastic cells are selected for initiation of tumors in Balb/c mice.
  • Histochemistry and Immunochemistry is performed on ascites and pleural effusion of patients with ovarian carcinoma to analyze structural characteristics of the neoplasia.
  • the markers that are monitored are cytokeratin(to identify ovarian neoplastic and mesothelial cells from inflammatory and mesenchymal cells); calretinin (to separate mesothelial from Her2/neu positive neoplastic cells); and CD45 (to separate inflammatory cells from the rest of the cell population in the samples). Additional markers that will be followed will include CD3 (T cells), CD20 (B cells), CD56 (NK cells), and CD 14 (monocytes).

Landscapes

  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention concerne des anticorps ou des fragments de ces anticorps qui se lient de manière spécifique au domaine extracellulaire de FcηRIIB, en particulier au FcηRIIB humain, et bloquent le site de liaison Fc de FcηRIIB. L'invention concerne également des procédés permettant de traiter le cancer et/ou de réguler l'activité cellulaire médiée par un complexe immunitaire par administration desdits anticorps destinés à renforcer une réponse immunitaire. L'invention concerne enfin des procédés permettant de rompre la tolérance à un antigène par administration d'un complexe antigène-anticorps et un anticorps selon l'invention.
PCT/US2005/045586 2004-12-15 2005-12-15 Anticorps specifiques de fc$g(g)riib et leurs procedes d'utilisation WO2006066078A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA2590935A CA2590935C (fr) 2004-12-15 2005-12-15 Anticorps specifiques de fc.gamma.riib et leurs procedes d'utilisation
EP05854332A EP1833849A4 (fr) 2004-12-15 2005-12-15 Anticorps specifiques de fc gamma riib et leurs procedes d'utilisation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63666304P 2004-12-15 2004-12-15
US60/636,663 2004-12-15

Publications (2)

Publication Number Publication Date
WO2006066078A2 true WO2006066078A2 (fr) 2006-06-22
WO2006066078A3 WO2006066078A3 (fr) 2006-10-19

Family

ID=36588589

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/045586 WO2006066078A2 (fr) 2004-12-15 2005-12-15 Anticorps specifiques de fc$g(g)riib et leurs procedes d'utilisation

Country Status (4)

Country Link
US (1) US20060177439A1 (fr)
EP (1) EP1833849A4 (fr)
CA (1) CA2590935C (fr)
WO (1) WO2006066078A2 (fr)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1747237A2 (fr) * 2004-04-16 2007-01-31 Macrogenics, Inc. Anticorps specifiques de gamma fc et methodes d'utilisation de ces anticorps
US7425620B2 (en) 2002-08-14 2008-09-16 Scott Koenig FcγRIIB-specific antibodies and methods of use thereof
EP2032159A2 (fr) * 2006-06-26 2009-03-11 Magrogenics, Inc. Combinaison d'anticorps de fc riib et d'anticorps spécifiques de cd20 et leurs procédés d'utilisation
US7521542B2 (en) 2004-05-10 2009-04-21 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
WO2009083009A2 (fr) * 2008-01-03 2009-07-09 Genmab A/S Anticorps monoclonaux anti-cd32b
US7655229B2 (en) 2004-09-02 2010-02-02 Chan Andrew C Anti-FC-gamma RIIB receptor antibody and uses therefor
US7662926B2 (en) 2004-09-02 2010-02-16 Genentech, Inc. Anti-Fc-gamma receptor antibodies, bispecific variants and uses therefor
WO2010033279A2 (fr) 2008-06-04 2010-03-25 Macrogenics, Inc. <sb>anticorps à liaison alteree à fcrn et leurs procedes d'utilisation</sb>
US8187593B2 (en) 2002-08-14 2012-05-29 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8697071B2 (en) 2005-08-10 2014-04-15 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
US9028815B2 (en) 2003-01-09 2015-05-12 Macrogenics, Inc. Identification and engineering of antibodies with variant FC regions and methods of using same
JP2016138129A (ja) * 2010-08-20 2016-08-04 ユニバーシティ、オブ、サウサンプトン Fcガンマriib(cd32b)特異的抗体およびcd20特異的抗体の併用使用
US9695236B2 (en) 2008-04-02 2017-07-04 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US9708408B2 (en) 2006-12-08 2017-07-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc Regions with altered affinities for FcγRactivating and FcγRinhibiting
US9714295B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
KR101799337B1 (ko) 2007-06-21 2017-12-20 마크로제닉스, 인크. 공유결합형 디아바디 및 이것의 사용
US9896508B2 (en) 2010-03-04 2018-02-20 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US10100116B2 (en) 2006-06-26 2018-10-16 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
US10131713B2 (en) 2008-04-02 2018-11-20 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
WO2019081983A1 (fr) 2017-10-25 2019-05-02 Novartis Ag Anticorps ciblant cd32b et leurs procédés d'utilisation
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8044180B2 (en) * 2002-08-14 2011-10-25 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8193318B2 (en) * 2002-08-14 2012-06-05 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
US8530627B2 (en) * 2002-08-14 2013-09-10 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2007024249A2 (fr) 2004-11-10 2007-03-01 Macrogenics, Inc. Fonction effectrice obtenue par creation par genie biologique de regions d'anticorps fc
EP1999470A4 (fr) * 2006-03-10 2009-08-19 Macrogenics Inc Identification et modification génétique d'anticorps avec des chaînes lourdes de variants et leurs procédés d'utilisation
WO2008105886A2 (fr) 2006-05-26 2008-09-04 Macrogenics, Inc. Anticorps spécifiques fcgriib humanisés, et leurs procédés d'utilisation
US20080112961A1 (en) * 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
EP2708557A1 (fr) 2007-05-30 2014-03-19 Xencor, Inc. Procédé et compositions pour inhiber des cellules exprimant CD32B
US8093018B2 (en) * 2008-05-20 2012-01-10 Otsuka Pharmaceutical Co., Ltd. Antibody identifying an antigen-bound antibody and an antigen-unbound antibody, and method for preparing the same
CN106432503B (zh) 2008-12-19 2020-03-06 宏观基因有限公司 共价双抗体及其用途
RU2583298C2 (ru) 2009-10-07 2016-05-10 Макродженикс, Инк. ПОЛИПЕПТИДЫ, СОДЕРЖАЩИЕ Fc-УЧАСТОК, КОТОРЫЕ ДЕМОНСТРИРУЮТ ПОВЫШЕННУЮ ЭФФЕКТОРНУЮ ФУНКЦИЮ БЛАГОДАРЯ ИЗМЕНЕНИЯМ СТЕПЕНИ ФУКОЗИЛИРОВАНИЯ, И СПОСОБЫ ИХ ПРИМЕНЕНИЯ
NZ618016A (en) 2011-05-21 2015-05-29 Macrogenics Inc Deimmunized serum-binding domains and their use for extending serum half-life
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
TW201709929A (zh) 2015-06-12 2017-03-16 宏觀基因股份有限公司 治療癌症的聯合療法
ES2963807T3 (es) 2016-06-08 2024-04-02 Xencor Inc Tratamiento de enfermedades relacionadas con la IgG4 con anticuerpos anti-CD19 de reticulación a CD32B
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
CN110172433B (zh) * 2019-05-12 2021-07-30 华中农业大学 一种产猪表皮生长因子的重组枯草芽孢杆菌工程菌及应用

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
CA2495251C (fr) * 2002-08-14 2018-03-06 Macrogenics, Inc. Anticorps specifiques du recepteur fc.gamma.riib et procedes d'utilisation de ces anticorps
US7355008B2 (en) * 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
MXPA06011796A (es) * 2004-04-16 2007-05-07 Macrogenics Inc Anticuerpos especificos de fc(riib y metodos para el uso de los mismos.
KR101297146B1 (ko) * 2004-05-10 2013-08-21 마크로제닉스, 인크. 인간화 FcγRIIB 특이적 항체 및 그의 사용 방법
ZA200701783B (en) * 2004-09-02 2009-10-28 Genentech Inc Anti-Fc-gamma RIIB receptor antibody and uses therefor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1833849A4 *

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7425620B2 (en) 2002-08-14 2008-09-16 Scott Koenig FcγRIIB-specific antibodies and methods of use thereof
US7425619B2 (en) 2002-08-14 2008-09-16 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8187593B2 (en) 2002-08-14 2012-05-29 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US9028815B2 (en) 2003-01-09 2015-05-12 Macrogenics, Inc. Identification and engineering of antibodies with variant FC regions and methods of using same
EP1747237A4 (fr) * 2004-04-16 2008-05-21 Macrogenics Inc Anticorps specifiques de gamma fc et methodes d'utilisation de ces anticorps
EP1747237A2 (fr) * 2004-04-16 2007-01-31 Macrogenics, Inc. Anticorps specifiques de gamma fc et methodes d'utilisation de ces anticorps
US7521542B2 (en) 2004-05-10 2009-04-21 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
US7662926B2 (en) 2004-09-02 2010-02-16 Genentech, Inc. Anti-Fc-gamma receptor antibodies, bispecific variants and uses therefor
US7655229B2 (en) 2004-09-02 2010-02-02 Chan Andrew C Anti-FC-gamma RIIB receptor antibody and uses therefor
US8697071B2 (en) 2005-08-10 2014-04-15 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US10100116B2 (en) 2006-06-26 2018-10-16 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
US9737599B2 (en) 2006-06-26 2017-08-22 Macrogenics, Inc. Combination of FcγRIIB-specific antibodies and CD20-specific antibodies and methods of use thereof
EP2032159A4 (fr) * 2006-06-26 2010-06-02 Macrogenics Inc Combinaison d'anticorps de fc riib et d'anticorps spécifiques de cd20 et leurs procédés d'utilisation
US11098125B2 (en) 2006-06-26 2021-08-24 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
EP2032159A2 (fr) * 2006-06-26 2009-03-11 Magrogenics, Inc. Combinaison d'anticorps de fc riib et d'anticorps spécifiques de cd20 et leurs procédés d'utilisation
US9708408B2 (en) 2006-12-08 2017-07-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc Regions with altered affinities for FcγRactivating and FcγRinhibiting
US10711069B2 (en) 2006-12-08 2020-07-14 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
US11787871B2 (en) 2006-12-08 2023-10-17 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having fc regions with altered affinities for FcgammaRactivating and FegammaRinhibiting
KR101799337B1 (ko) 2007-06-21 2017-12-20 마크로제닉스, 인크. 공유결합형 디아바디 및 이것의 사용
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
US20110038869A1 (en) * 2008-01-03 2011-02-17 Genmab A/S Monoclonal antibodies against cd32b
US8802089B2 (en) 2008-01-03 2014-08-12 Genmab A/S Monoclonal antibodies against CD32B
WO2009083009A2 (fr) * 2008-01-03 2009-07-09 Genmab A/S Anticorps monoclonaux anti-cd32b
US9663578B2 (en) 2008-01-03 2017-05-30 Genmab A/S Monoclonal antibodies against CD32b
WO2009083009A3 (fr) * 2008-01-03 2009-08-27 Genmab A/S Anticorps monoclonaux anti-cd32b
US20150004177A1 (en) * 2008-01-03 2015-01-01 Genmab A/S Monoclonal antibodies against cd32b
US9695236B2 (en) 2008-04-02 2017-07-04 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US10479831B2 (en) 2008-04-02 2019-11-19 Macrogenics, Inc BCR-complex-specific antibodies and methods of using same
US11028183B2 (en) 2008-04-02 2021-06-08 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US10131713B2 (en) 2008-04-02 2018-11-20 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
WO2010033279A2 (fr) 2008-06-04 2010-03-25 Macrogenics, Inc. <sb>anticorps à liaison alteree à fcrn et leurs procedes d'utilisation</sb>
US9714295B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US10683364B2 (en) 2010-03-04 2020-06-16 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US10730945B2 (en) 2010-03-04 2020-08-04 Macrogenics, Inc. Antibodies reactive with B7-H3 and users thereof
US9896508B2 (en) 2010-03-04 2018-02-20 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US9714296B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US10525129B2 (en) 2010-08-20 2020-01-07 University Of Southampton Combined use of Fc gamma RIIb (CD32B) and CD20-specific antibodies
CN114099669A (zh) * 2010-08-20 2022-03-01 南安普敦大学 FcγRIIB(CD32B)和CD20特异性抗体的组合应用
CN114099666A (zh) * 2010-08-20 2022-03-01 南安普敦大学 FcγRIIB(CD32B)和CD20特异性抗体的组合应用
US11623005B2 (en) 2010-08-20 2023-04-11 University Of Southampton Combined use of FcγRIIb (CD32B) and CD20 specific antibodies
JP2016138129A (ja) * 2010-08-20 2016-08-04 ユニバーシティ、オブ、サウサンプトン Fcガンマriib(cd32b)特異的抗体およびcd20特異的抗体の併用使用
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US11591400B2 (en) 2016-04-15 2023-02-28 Macrogenics, Inc. B7-H3 directed antibody drug conjugates
WO2019081983A1 (fr) 2017-10-25 2019-05-02 Novartis Ag Anticorps ciblant cd32b et leurs procédés d'utilisation

Also Published As

Publication number Publication date
WO2006066078A3 (fr) 2006-10-19
CA2590935C (fr) 2014-09-30
CA2590935A1 (fr) 2006-06-22
EP1833849A4 (fr) 2009-08-05
US20060177439A1 (en) 2006-08-10
EP1833849A2 (fr) 2007-09-19

Similar Documents

Publication Publication Date Title
US20220177574A1 (en) FcgammaRIIB-Specific Antibodies and Methods of Use Thereof
CA2590935C (fr) Anticorps specifiques de fc.gamma.riib et leurs procedes d&#39;utilisation
US7425620B2 (en) FcγRIIB-specific antibodies and methods of use thereof
AU2005247301B2 (en) FC&amp;gamma;RIIB-specific antibodies and methods of use thereof
US8044180B2 (en) FcγRIIB specific antibodies and methods of use thereof
US8968730B2 (en) FcγRIIB specific antibodies and methods of use thereof
US8530627B2 (en) FcγRIIB specific antibodies and methods of use thereof
US20060013810A1 (en) Humanized FcgammaRIIB-specific antibodies and methods of use thereof
WO2005018669A1 (fr) Anticorps specifiques des proteines fc?riib et procedes d&#39;utilisation associes
US20150344574A1 (en) FcGammaRIIB Specific Antibodies and Methods of Use Thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2590935

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005854332

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2005854332

Country of ref document: EP